1
|
Byer-Alcorace A, Thomas C, Taub ME, Piekos S. Improved clearance predictions for aldehyde oxidase substrates using a novel triculture human hepatocyte model. Drug Metab Dispos 2025; 53:100051. [PMID: 40147225 DOI: 10.1016/j.dmd.2025.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 03/29/2025] Open
Abstract
Over the last several decades, efforts in medicinal chemistry have aimed to reduce the extent of CYP metabolism of new chemical entities. This approach, however, has led to increased susceptibility to metabolism by non-CYP-mediated pathways, particularly involving other phase I enzymes such as aldehyde oxidase (AO). Commonly used in vitro models, such as suspended or cocultured primary human hepatocytes, have limitations in evaluating the disposition of compounds metabolized by AO due to low or variable levels of enzyme activity. Thus, an in vitro model that exhibits high to moderate levels of AO activity that can better predict the contribution of AO to drug metabolism and its impact on drug clearance is needed. A novel, 2D+ primary human hepatocyte model, TruVivo, was evaluated for its potential utility to improve hepatic clearance (CLh) predictions and determine the contribution of AO to drug metabolism in humans. TruVivo demonstrated stable levels of AO activity for at least 2 weeks that were higher than levels in other hepatocyte models. CLh predictions generated using TruVivo for the reference compounds carbazeran, zoniporide, zaleplon, and O6-benzylguanine were within 2-fold of reported in vivo CL values. Furthermore, the estimated fraction metabolized by AO for zaleplon and zoniporide was within 25% of reported in vivo values, whereas that for carbazeran and O6-benzylguanine was similar to those generated in other systems. These findings suggest TruVivo may offer a novel means to assess CLh of AO substrates more accurately, even over extended incubation times for low clearance compounds. SIGNIFICANCE STATEMENT: The TruVivo in vitro primary human hepatocyte model maintains high levels of aldehyde oxidase (AO) activity for at least 14 days in culture, making the system suitable for evaluating slowly metabolized compounds, particularly those metabolized by AO. This novel system may therefore be useful for improving human clearance predictions for AO substrates.
Collapse
Affiliation(s)
- Alexander Byer-Alcorace
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Cody Thomas
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut
| | - Stephanie Piekos
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, Connecticut.
| |
Collapse
|
2
|
Subash S, Singh DK, Ahire D, Khojasteh SC, Murray BP, Zientek MA, Jones RS, Kulkarni P, Zubair F, Smith BJ, Heyward S, Leeder JS, Prasad B. Ontogeny of Human Liver Aldehyde Oxidase: Developmental Changes and Implications for Drug Metabolism. Mol Pharm 2024; 21:2740-2750. [PMID: 38717252 DOI: 10.1021/acs.molpharmaceut.3c01147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Despite the increasing importance of aldehyde oxidase (AO) in the drug metabolism of clinical candidates, ontogeny data for AO are limited. The objective of our study was to characterize the age-dependent AO content and activity in the human liver cytosolic fraction (HLC) and human hepatocytes (HH). HLC (n = 121 donors) and HH (n = 50 donors) were analyzed for (1) AO protein content by quantitative proteomics and (2) enzyme activity using carbazeran as a probe substrate. AO activity showed high technical variability and poor correlation with the content in HLC samples, whereas hepatocyte samples showed a strong correlation between the content and activity. Similarly, AO content and activity showed no significant age-dependent differences in HLC samples, whereas the average AO content and activity in hepatocytes increased significantly (∼20-40-fold) from the neonatal levels (0-28 days). Based on the hepatocyte data, the age at which 50% of the adult AO content is reached (age50) was 3.15 years (0.32-13.97 years, 95% CI). Metabolite profiling of carbazeran revealed age-dependent metabolic switching and the role of non-AO mechanisms (glucuronidation and desmethylation) in carbazeran elimination. The content-activity correlation in hepatocytes improved significantly (R2 = 0.95; p < 0.0001) in samples showing <10% contribution of glucuronidation toward the overall metabolism, confirming that AO-mediated oxidation and glucuronidation are the key routes of carbazeran metabolism. Considering the confounding effect of glucuronidation on AO activity, AO content-based ontogeny data are a more direct reflection of developmental changes in protein expression. The comprehensive ontogeny data of AO in HH samples are more reliable than HLC data, which are important for developing robust physiologically based pharmacokinetic models for predicting AO-mediated metabolism in children.
Collapse
Affiliation(s)
- Sandhya Subash
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - Dilip K Singh
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - Deepak Ahire
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| | - S Cyrus Khojasteh
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Bernard P Murray
- Drug Metabolism, Gilead Sciences, Inc., Foster City, California 94404, United States
| | - Michael A Zientek
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Robert S Jones
- Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California 94080, United States
| | - Priyanka Kulkarni
- Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals, Inc., Cambridge, Massachusetts 02139, United States
| | - Faizan Zubair
- Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc., San Diego, California 92121, United States
| | - Bill J Smith
- Terminal Phase Consulting LLC, Colorado Springs, Colorado 94404, United States
| | - Scott Heyward
- BioIVT, Inc., Baltimore, Maryland 21227, United States
| | - J Steven Leeder
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri 64108, United States
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington 99202, United States
| |
Collapse
|
3
|
Yu M, Zhu ZF, Yang F, Yuan YF, Liao SD, Liu ML, Cheng X. Different Anti-inflammatory Drugs on High-Sensitivity C-Reactive Protein in Patients After Percutaneous Coronary Intervention: A Pilot Randomized Clinical Trial. J Cardiovasc Pharmacol 2024; 83:234-242. [PMID: 37944130 DOI: 10.1097/fjc.0000000000001509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 10/04/2023] [Indexed: 11/12/2023]
Abstract
ABSTRACT Colchicine reduces atherothrombotic cardiovascular events in coronary artery disease because of its anti-inflammatory effect. However, the effects of the other anti-inflammatory drugs in coronary artery disease remain unclear. This study included 132 patients aged 18-80 years who completed the planned percutaneous coronary interventions and were treated with aggressive secondary prevention strategies for 4 weeks. The subjects were randomly assigned to 1 of the following treatment groups for 4 weeks: (1) control: no additional intervention; (2) colchicine: 0.5 mg once a day; (3) tranilast: 0.1 g thrice a day; or (4) oridonin: 0.5 g thrice a day. The primary outcome was the percentage change in high-sensitivity C-reactive protein (hsCRP) levels at the end of 4 weeks. In total, 109 patients completed the study. The mean age was 58.33 years, 81 (74.31%) were male, and 28 (25.69%) were female. The percentage changes in hsCRP after 4 weeks of treatment were -11.62%, -48.28%, -21.60%, and -7.81%, in the control, colchicine, tranilast, and the oridonin groups, respectively. Compared with the control group, only the colchicine group showed significantly greater reduction in hsCRP levels ( P = 0.022). In targeted proteomic analysis, proteins associated with neutrophil activation (azurocidin, myeloperoxidase, and myeloblastin), platelet aggregation (glycoprotein VI), and endothelial damage (galectin-3) were reduced with colchicine therapy. These results show that of 3 anti-inflammatory drugs only colchicine could reduce hsCRP in patients after percutaneous coronary interventions.
Collapse
Affiliation(s)
- Miao Yu
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng-Feng Zhu
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Yang
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan-Fan Yuan
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu-De Liao
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mei-Lin Liu
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Cheng
- Department of Cardiology
- Hubei Key Laboratory of Biological Targeted Therapy; and
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Abramenko N, Vellieux F, Veselá K, Kejík Z, Hajduch J, Masařík M, Babula P, Hoskovec D, Pacák K, Martásek P, Smetana K, Jakubek M. Investigation of the potential effects of estrogen receptor modulators on immune checkpoint molecules. Sci Rep 2024; 14:3043. [PMID: 38321096 PMCID: PMC10847107 DOI: 10.1038/s41598-024-51804-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024] Open
Abstract
Immune checkpoints regulate the immune system response. Recent studies suggest that flavonoids, known as phytoestrogens, may inhibit the PD-1/PD-L1 axis. We explored the potential of estrogens and 17 Selective Estrogen Receptor Modulators (SERMs) as inhibiting ligands for immune checkpoint proteins (CTLA-4, PD-L1, PD-1, and CD80). Our docking studies revealed strong binding energy values for quinestrol, quercetin, and bazedoxifene, indicating their potential to inhibit PD-1 and CTLA-4. Quercetin and bazedoxifene, known to modulate EGFR and IL-6R alongside estrogen receptors, can influence the immune checkpoint functionality. We discuss the impact of SERMs on PD-1 and CTLA-4, suggesting that these SERMs could have therapeutic effects through immune checkpoint inhibition. This study highlights the potential of SERMs as inhibitory ligands for immune checkpoint proteins, emphasizing the importance of considering PD-1 and CTLA-4 inhibition when evaluating SERMs as therapeutic agents. Our findings open new avenues for cancer immunotherapy by exploring the interaction between various SERMs and immune checkpoint pathways.
Collapse
Affiliation(s)
- Nikita Abramenko
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Fréderic Vellieux
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Jan Hajduch
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
| | - Michal Masařík
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, 121 08, Prague, Czech Republic
| | - Karel Pacák
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Building 10, Room 1-3140, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic
| | - Karel Smetana
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic
- Institute of Anatomy, First Faculty of Medicine, Charles University, 120 00, Prague, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, 252 50, Vestec, Czech Republic.
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, 120 00, Prague, Czech Republic.
| |
Collapse
|
5
|
Hammid A, Fallon JK, Vellonen KS, Lassila T, Reinisalo M, Urtti A, Gonzalez F, Tolonen A, Smith PC, Honkakoski P. Aldehyde oxidase 1 activity and protein expression in human, rabbit, and pig ocular tissues. Eur J Pharm Sci 2023; 191:106603. [PMID: 37827455 DOI: 10.1016/j.ejps.2023.106603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/18/2023] [Accepted: 10/06/2023] [Indexed: 10/14/2023]
Abstract
Aldehyde oxidase (AOX) is a cytosolic drug-metabolizing enzyme which has attracted increasing attention in drug development due to its high hepatic expression, broad substrate profile and species differences. In contrast, there is limited information on the presence and activity of AOX in extrahepatic tissues including ocular tissues. Because several ocular drugs are potential substrates for AOX, we performed a comprehensive analysis of the AOX1 expression and activity profile in seven ocular tissues from humans, rabbits, and pigs. AOX activities were determined using optimized assays for the established human AOX1 probe substrates 4-dimethylamino-cinnamaldehyde (DMAC) and phthalazine. Inhibition studies were undertaken in conjunctival and retinal homogenates using well-established human AOX1 inhibitors menadione and chlorpromazine. AOX1 protein contents were quantitated with targeted proteomics and confirmed by immunoblotting. Overall, DMAC oxidation rates varied over 10-fold between species (human ˃˃ rabbit ˃ pig) and showed 2- to 6-fold differences between tissues from the same species. Menadione seemed a more potent inhibitor of DMAC oxidation across species than chlorpromazine. Human AOX1 protein levels were highest in the conjunctiva, followed by most posterior tissues, whereas anterior tissues showed low levels. The rabbit AOX1 expression was high in the conjunctiva, retinal pigment epithelial (RPE), and choroid while lower in the anterior tissues. Quantification of pig AOX1 was not successful but immunoblotting confirmed the presence of AOX1 in all species. DMAC oxidation rates and AOX1 contents correlated quite well in humans and rabbits. This study provides, for the first time, insights into the ocular expression and activity of AOX1 among multiple species.
Collapse
Affiliation(s)
- Anam Hammid
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, FI-70210 Kuopio, Finland.
| | - John K Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Campus Box 7355, Chapel Hill, NC 27599-7355, United States
| | - Kati-Sisko Vellonen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, FI-70210 Kuopio, Finland
| | - Toni Lassila
- Admescope Ltd, Typpitie 1, FI-90620 Oulu, Finland
| | - Mika Reinisalo
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, FI-70210 Kuopio, Finland
| | - Arto Urtti
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, FI-70210 Kuopio, Finland; Faculty of Pharmacy, University of Helsinki, Viikinkaari 5 E, FI-00790 Helsinki, Finland
| | - Francisco Gonzalez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Service of Ophthalmology, University Hospital of Santiago de Compostela, and Fundacion Instituto de Investigacion Sanitaria de Santiago de Compostela (FIDIS), 15706 Santiago de Compostela, Spain
| | - Ari Tolonen
- Admescope Ltd, Typpitie 1, FI-90620 Oulu, Finland
| | - Philip C Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Campus Box 7355, Chapel Hill, NC 27599-7355, United States
| | - Paavo Honkakoski
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1 C, FI-70210 Kuopio, Finland
| |
Collapse
|
6
|
Izat N, Bolleddula J, Abbasi A, Cheruzel L, Jones RS, Moss D, Ortega-Muro F, Parmentier Y, Peterkin VC, Tian DD, Venkatakrishnan K, Zientek MA, Barber J, Houston JB, Galetin A, Scotcher D. Challenges and Opportunities for In Vitro-In Vivo Extrapolation of Aldehyde Oxidase-Mediated Clearance: Toward a Roadmap for Quantitative Translation. Drug Metab Dispos 2023; 51:1591-1606. [PMID: 37751998 DOI: 10.1124/dmd.123.001436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Underestimation of aldehyde oxidase (AO)-mediated clearance by current in vitro assays leads to uncertainty in human dose projections, thereby reducing the likelihood of success in drug development. In the present study we first evaluated the current drug development practices for AO substrates. Next, the overall predictive performance of in vitro-in vivo extrapolation of unbound hepatic intrinsic clearance (CLint,u) and unbound hepatic intrinsic clearance by AO (CLint,u,AO) was assessed using a comprehensive literature database of in vitro (human cytosol/S9/hepatocytes) and in vivo (intravenous/oral) data collated for 22 AO substrates (total of 100 datapoints from multiple studies). Correction for unbound fraction in the incubation was done by experimental data or in silico predictions. The fraction metabolized by AO (fmAO) determined via in vitro/in vivo approaches was found to be highly variable. The geometric mean fold errors (gmfe) for scaled CLint,u (mL/min/kg) were 10.4 for human hepatocytes, 5.6 for human liver cytosols, and 5.0 for human liver S9, respectively. Application of these gmfe's as empirical scaling factors improved predictions (45%-57% within twofold of observed) compared with no correction (11%-27% within twofold), with the scaling factors qualified by leave-one-out cross-validation. A road map for quantitative translation was then proposed following a critical evaluation on the in vitro and clinical methodology to estimate in vivo fmAO In conclusion, the study provides the most robust system-specific empirical scaling factors to date as a pragmatic approach for the prediction of in vivo CLint,u,AO in the early stages of drug development. SIGNIFICANCE STATEMENT: Confidence remains low when predicting in vivo clearance of AO substrates using in vitro systems, leading to de-prioritization of AO substrates from the drug development pipeline to mitigate risk of unexpected and costly in vivo impact. The current study establishes a set of empirical scaling factors as a pragmatic tool to improve predictability of in vivo AO clearance. Developing clinical pharmacology strategies for AO substrates by utilizing mass balance/clinical drug-drug interaction data will help build confidence in fmAO.
Collapse
Affiliation(s)
- Nihan Izat
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jayaprakasam Bolleddula
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Armina Abbasi
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Lionel Cheruzel
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Robert S Jones
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Darren Moss
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Fatima Ortega-Muro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Yannick Parmentier
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Vincent C Peterkin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Dan-Dan Tian
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Karthik Venkatakrishnan
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Michael A Zientek
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester, UK (N.I., Ji.B., J.B.H., A.G., D.S.); EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (Ja.B., K.V.); Amgen Inc., South San Francisco, California (A.A.); Genentech, Inc., South San Francisco, California (L.C., R.S.J.); Janssen Pharmaceutical Companies of Johnson & Johnson, Beerse, Belgium (D.M.); GSK R&D, Tres Cantos, Madrid, Spain (F.O.M.); Technologie Servier, Orléans, France (Y.P.); AbbVie Inc., North Chicago, Illinois (V.C.P.); Eli Lilly and Company, Indianapolis, Indiana (D.-D.T.); and Takeda Pharmaceuticals Limited, San Diego, California (M.A.Z.)
| |
Collapse
|
7
|
Subash S, Singh DK, Ahire DS, Khojasteh SC, Murray BP, Zientek MA, Jones RS, Kulkarni P, Smith BJ, Heyward S, Cronin CN, Prasad B. Dissecting Parameters Contributing to the Underprediction of Aldehyde Oxidase-Mediated Metabolic Clearance of Drugs. Drug Metab Dispos 2023; 51:1362-1371. [PMID: 37429730 DOI: 10.1124/dmd.123.001379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/01/2023] [Accepted: 07/07/2023] [Indexed: 07/12/2023] Open
Abstract
We investigated the effect of variability and instability in aldehyde oxidase (AO) content and activity on the scaling of in vitro metabolism data. AO content and activity in human liver cytosol (HLC) and five recombinant human AO preparations (rAO) were determined using targeted proteomics and carbazeran oxidation assay, respectively. AO content was highly variable as indicated by the relative expression factor (REF; i.e., HLC to rAO content) ranging from 0.001 to 1.7 across different in vitro systems. The activity of AO in HLC degrades at a 10-fold higher rate in the presence of the substrate as compared with the activity performed after preincubation without substrate. To scale the metabolic activity from rAO to HLC, a protein-normalized activity factor (pnAF) was proposed wherein the activity was corrected by AO content, which revealed up to sixfold higher AO activity in HLC versus rAO systems. A similar value of pnAF was observed for another substrate, ripasudil. Physiologically based pharmacokinetic (PBPK) modeling revealed a significant additional clearance (CL; 66%), which allowed for the successful prediction of in vivo CL of four other substrates, i.e., O-benzyl guanine, BIBX1382, zaleplon, and zoniporide. For carbazeran, the metabolite identification study showed that the direct glucuronidation may be contributing to around 12% elimination. Taken together, this study identified differential protein content, instability of in vitro activity, role of additional AO clearance, and unaccounted metabolic pathways as plausible reasons for the underprediction of AO-mediated drug metabolism. Consideration of these factors and integration of REF and pnAF in PBPK models will allow better prediction of AO metabolism. SIGNIFICANCE STATEMENT: This study elucidated the plausible reasons for the underprediction of aldehyde oxidase (AO)-mediated drug metabolism and provided recommendations to address them. It demonstrated that integrating protein content and activity differences and accounting for the loss of AO activity, as well as consideration of extrahepatic clearance and additional pathways, would improve the in vitro to in vivo extrapolation of AO-mediated drug metabolism using physiologically based pharmacokinetic modeling.
Collapse
Affiliation(s)
- Sandhya Subash
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Dilip K Singh
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Deepak S Ahire
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - S Cyrus Khojasteh
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bernard P Murray
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Michael A Zientek
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Robert S Jones
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Priyanka Kulkarni
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bill J Smith
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Ciarán N Cronin
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University (WSU), Spokane, Washington (S.S., D.K.S., D.S.A., B.P.); Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (S.C.K., R.S.J.); Drug Metabolism, Gilead Sciences, Foster City, California (B.P.M., B.J.S.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, San Diego, California (M.A.Z.); Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Cambridge, Massachusetts (P.K.); BioIVT Inc., Baltimore, Maryland (S.H.); and Structural Biology and Protein Sciences, Pfizer Global Research & Development and Medical, La Jolla, California (C.N.C.)
| |
Collapse
|
8
|
Inoue K, Kikuchi K, Takahashi K, Hitaoka S, Kusano K, Komori T. In Vitro Metabolism of 2-Methoxy-N-[3-[4-[3-methyl-4-[(6-methyl- 3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) by Aldehyde Oxidase and Predicting Its Percent Contribution Relative to CYP-Mediated Metabolism. Drug Metab Dispos 2023; 51:962-969. [PMID: 37188528 DOI: 10.1124/dmd.122.000995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 02/19/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) is an anticancer drug that was discontinued due to hepatotoxicity found in clinical studies. Metabolite analysis of CP-724,714 was conducted using human hepatocytes, in which twelve oxidative metabolites and one hydrolyzed metabolite were formed. Among the three mono-oxidative metabolites, the formation of two was inhibited by adding 1-aminobenzotriazole, a pan-CYP inhibitor. In contrast, the remaining one was not affected by this inhibitor but partially inhibited by hydralazine, indicating that aldehyde oxidase (AO) was involved in metabolizing CP-724,714, which contains a quinazoline substructure, a heterocyclic aromatic quinazoline ring, known to be preferably metabolized by AO. One of the oxidative metabolites of CP-724,714 observed in human hepatocytes was also generated in recombinant human AO. Although CP-724,714 is metabolized by both CYPs and AO in human hepatocytes, the contribution level of AO could not be evaluated using its specific inhibitors because of low AO activity in in vitro human materials. Here, we present a metabolic pathway for CP-724,714 in human hepatocytes and the involvement of AO in CP-724,714 metabolism. We showed here a plausible workflow for predicting AO contribution to the metabolism of CP-724,714 based on DMPK screening data. SIGNIFICANCE STATEMENT: 2-methoxy-N-[3-[4-[3-methyl-4-[(6-methyl-3-pyridinyl)oxy]anilino]-6-quinazolinyl]prop-2-enyl]acetamide (CP-724,714) was identified as a substrate of aldehyde oxidase (AO) rather than xanthine oxidase. Since CP-724,714 is also metabolized by cytochrome P450s (CYPs), the contribution levels of AO and CYPs in the metabolism of CP-724,714 were estimated simultaneously based on in vitro drug metabolism screening data.
Collapse
Affiliation(s)
- Kazuko Inoue
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kiyomi Kikuchi
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kazumi Takahashi
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Seiji Hitaoka
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Kazutomi Kusano
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| | - Takafumi Komori
- Global Drug Metabolism and Pharmacokinetics, Biopharmaceutical Assessments Unit, DHBL, Eisai Co., Ltd., Tsukuba, Japan, (K.I., K.K., K.T., Ka.K., T.K.) and Emerging Modality Generation Department, Discovery Evidence Generation Function, DHBL, Eisai Co., Ltd., Tsukuba, Japan (S.H.)
| |
Collapse
|
9
|
Esmaeeli M, Nimtz M, Jänsch L, Ruddock LW, Leimkühler S. Inactivation of Human Aldehyde Oxidase by Small Sulfhydryl-Containing Reducing Agents. Drug Metab Dispos 2023; 51:764-770. [PMID: 37012073 DOI: 10.1124/dmd.122.001244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/13/2023] [Accepted: 03/07/2023] [Indexed: 04/05/2023] Open
Abstract
Human aldehyde oxidase (hAOX1) is a molybdoflavoenzyme that belongs to the xanthine oxidase (XO) family. hAOX1 is involved in phase I drug metabolism, but its physiologic role is not fully understood to date, and preclinical studies consistently underestimated hAOX1 clearance. In the present work, we report an unexpected effect of the common sulfhydryl-containing reducing agents, e.g., dithiothreitol (DTT), on the activity of hAOX1 and mouse aldehyde oxidases. We demonstrate that this effect is due to the reactivity of the sulfido ligand bound at the molybdenum cofactor with the sulfhydryl groups. The sulfido ligand coordinated to the Mo atom in the XO family of enzymes plays a crucial role in the catalytic cycle and its removal results in the total inactivation of these enzymes. Because liver cytosols, S9 fractions, and hepatocytes are commonly used to screen the drug candidates for hAOX1, our study suggests that DTT treatment of these samples should be avoided, otherwise false negative results by an inactivated hAOX1 might be obtained. SIGNIFICANCE STATEMENT: This work characterizes the inactivation of human aldehyde oxidase (hAOX1) by sulfhydryl-containing agents and identifies the site of inactivation. The role of dithiothreitol in the inhibition of hAOX1 should be considered for the preparation of hAOX1-containing fractions for pharmacological studies on drug metabolism and drug clearance.
Collapse
Affiliation(s)
- Mariam Esmaeeli
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Manfred Nimtz
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Lothar Jänsch
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Lloyd W Ruddock
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| | - Silke Leimkühler
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany (M.E., S.L.); Helmholtz Center for Infection Research, Inhoffenstraße 7, Braunschweig, Germany (M.N., L.J.); and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie A, Oulu, Finland (L.W.R.)
| |
Collapse
|
10
|
Choi C, Carlo AA, Cronin CN, Jing K, Kung DW, Liu J, Lombardo VM, Turco AR, Yin J, Yu A, Wright SW. Synthesis of Deuterated Heterocycles with Me 2NCD(OMe) 2 and Evaluation of the Products for Metabolism by Aldehyde Oxidase. ACS Med Chem Lett 2022; 13:250-256. [PMID: 35178182 PMCID: PMC8842100 DOI: 10.1021/acsmedchemlett.1c00634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
The metabolic oxidation of drug-like small molecules by aldehyde oxidase (AO) has commonly been mitigated through the incorporation of deuterium at the oxidation site. We report that dimethylformamide dimethyl acetal and related compounds undergo rapid CH to CD isotopic exchange upon exposure to methanol-d and similar deuterated alcohols. This isotopic exchange process can be used to synthesize Me2NCD(OMe)2 and has significant implications for the use of Me2NCD(OMe)2 in the synthesis of specifically deuterium-labeled compounds. The application of Me2NCD(OMe)2 to the synthesis of various heterocycles that have been associated with AO metabolism is described, and we report the impact of deuteration on the rate of in vitro AO-mediated metabolism.
Collapse
Affiliation(s)
- Chulho Choi
- Medicine
Design, Pfizer Worldwide Research, Development
and Medical, Eastern Point Road, Groton, Connecticut 06340, United States,
| | - Anthony A. Carlo
- Structural
Biology and Protein Sciences, Pfizer Worldwide
Research, Development and Medical, La Jolla, California 92121, United States
| | - Ciarán N. Cronin
- Structural
Biology and Protein Sciences, Pfizer Worldwide
Research, Development and Medical, La Jolla, California 92121, United States
| | - Konghua Jing
- WuXi
AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Daniel W. Kung
- Medicine
Design, Pfizer Worldwide Research, Development
and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Jianhua Liu
- Structural
Biology and Protein Sciences, Pfizer Worldwide
Research, Development and Medical, La Jolla, California 92121, United States
| | - Vincent M. Lombardo
- Medicine
Design, Pfizer Worldwide Research, Development
and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Abigail R. Turco
- Medicine
Design, Pfizer Worldwide Research, Development
and Medical, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Juxing Yin
- WuXi
AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Aijia Yu
- WuXi
AppTec, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China
| | - Stephen W. Wright
- Medicine
Design, Pfizer Worldwide Research, Development
and Medical, Eastern Point Road, Groton, Connecticut 06340, United States,
| |
Collapse
|
11
|
Chitosan for biomedical applications, promising antidiabetic drug delivery system, and new diabetes mellitus treatment based on stem cell. Int J Biol Macromol 2021; 190:417-432. [PMID: 34450151 DOI: 10.1016/j.ijbiomac.2021.08.154] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/14/2021] [Accepted: 08/18/2021] [Indexed: 02/07/2023]
Abstract
Since chitosan's excellent pharmacokinetic and chemical properties, it is an attractive and promising carbohydrate biopolymer in biomedical applications. Chitosan's beneficial function in the defense and propagation of pancreatic β cells, reducing hyperglycemia, and avoiding diabetes mellitus associated with impaired lipid metabolism has been demonstrated in several studies. Additionally, chitosan has also been used in various nanocarriers to deliver various antidiabetic drugs to reduce glucose levels. Herein, the first to provide the currently available potential benefits of chitosan in diabetes mellitus treatment focuses on chitosan-based nanocarriers for oral administration of various antidiabetic drugs nasal and subcutaneous passages. Moreover, chitosan is used to activate and deliver stem cells and differentiate them into cells similar to pancreatic beta cells as a new type of treatment for type one diabetes mellitus. The results of this review will be helpful in the development of promising treatments and better control of diabetes mellitus.
Collapse
|
12
|
Pharmacokinetic/Pharmacodynamic Modelling of Allopurinol, its Active Metabolite Oxypurinol, and Biomarkers Hypoxanthine, Xanthine and Uric Acid in Hypoxic-Ischemic Encephalopathy Neonates. Clin Pharmacokinet 2021; 61:321-333. [PMID: 34617261 PMCID: PMC8813842 DOI: 10.1007/s40262-021-01068-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2021] [Indexed: 12/04/2022]
Abstract
Background Allopurinol, an xanthine oxidase (XO) inhibitor, is a promising intervention that may provide neuroprotection for neonates with hypoxic-ischemic encephalopathy (HIE). Currently, a double-blind, placebo-controlled study (ALBINO, NCT03162653) is investigating the neuroprotective effect of allopurinol in HIE neonates. Objective The aim of the current study was to establish the pharmacokinetics (PK) of allopurinol and oxypurinol, and the pharmacodynamics (PD) of both compounds on hypoxanthine, xanthine, and uric acid in HIE neonates. The dosage used and the effect of allopurinol in this population, either or not undergoing therapeutic hypothermia (TH), were evaluated. Methods Forty-six neonates from the ALBINO study and two historical clinical studies were included. All doses were administered on the first day of life. In the ALBINO study (n = 20), neonates received a first dose of allopurinol 20 mg/kg, and, in the case of TH (n = 13), a second dose of allopurinol 10 mg/kg. In the historical cohorts (n = 26), neonates (all without TH) received two doses of allopurinol 20 mg/kg in total. Allopurinol and oxypurinol population PK, and their effects on inhibiting conversions of hypoxanthine and xanthine to uric acid, were assessed using nonlinear mixed-effects modelling. Results Allopurinol and oxypurinol PK were described by two sequential one-compartment models with an autoinhibition effect on allopurinol metabolism by oxypurinol. For allopurinol, clearance (CL) was 0.83 L/h (95% confidence interval [CI] 0.62–1.09) and volume of distribution (Vd) was 2.43 L (95% CI 2.25–2.63). For metabolite oxypurinol, CL and Vd relative to a formation fraction (fm) were 0.26 L/h (95% CI 0.23–0.3) and 11 L (95% CI 9.9–12.2), respectively. No difference in allopurinol and oxypurinol CL was found between TH and non-TH patients. The effect of allopurinol and oxypurinol on XO inhibition was described by a turnover model of hypoxanthine with sequential metabolites xanthine and uric acid. The combined allopurinol and oxypurinol concentration at the half-maximal XO inhibition was 0.36 mg/L (95% CI 0.31–0.42). Conclusion The PK and PD of allopurinol, oxypurinol, hypoxanthine, xanthine, and uric acid in neonates with HIE were described. The dosing regimen applied in the ALBINO trial leads to the targeted XO inhibition in neonates treated with or without TH. Supplementary Information The online version contains supplementary material available at 10.1007/s40262-021-01068-0.
Collapse
|
13
|
Mota C, Diniz A, Coelho C, Santos-Silva T, Esmaeeli M, Leimkühler S, Cabrita EJ, Marcelo F, Romão MJ. Interrogating the Inhibition Mechanisms of Human Aldehyde Oxidase by X-ray Crystallography and NMR Spectroscopy: The Raloxifene Case. J Med Chem 2021; 64:13025-13037. [PMID: 34415167 DOI: 10.1021/acs.jmedchem.1c01125] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Human aldehyde oxidase (hAOX1) is mainly present in the liver and has an emerging role in drug metabolism, since it accepts a wide range of molecules as substrates and inhibitors. Herein, we employed an integrative approach by combining NMR, X-ray crystallography, and enzyme inhibition kinetics to understand the inhibition modes of three hAOX1 inhibitors-thioridazine, benzamidine, and raloxifene. These integrative data indicate that thioridazine is a noncompetitive inhibitor, while benzamidine presents a mixed type of inhibition. Additionally, we describe the first crystal structure of hAOX1 in complex with raloxifene. Raloxifene binds tightly at the entrance of the substrate tunnel, stabilizing the flexible entrance gates and elucidating an unusual substrate-dependent mechanism of inhibition with potential impact on drug-drug interactions. This study can be considered as a proof-of-concept for an efficient experimental screening of prospective substrates and inhibitors of hAOX1 relevant in drug discovery.
Collapse
Affiliation(s)
- Cristiano Mota
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Ana Diniz
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Catarina Coelho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Teresa Santos-Silva
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Mariam Esmaeeli
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Silke Leimkühler
- Department of Molecular Enzymology, Institute of Biochemistry and Biology, University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Eurico J Cabrita
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Filipa Marcelo
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Maria João Romão
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal.,UCIBIO-Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| |
Collapse
|
14
|
Garrido C, Leimkühler S. The Inactivation of Human Aldehyde Oxidase 1 by Hydrogen Peroxide and Superoxide. Drug Metab Dispos 2021; 49:729-735. [PMID: 34183377 DOI: 10.1124/dmd.121.000549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/21/2021] [Indexed: 12/14/2022] Open
Abstract
Mammalian aldehyde oxidases (AOX) are molybdo-flavoenzymes of pharmacological and pathophysiologic relevance that are involved in phase I drug metabolism and, as a product of their enzymatic activity, are also involved in the generation of reactive oxygen species. So far, the physiologic role of aldehyde oxidase 1 in the human body remains unknown. The human enzyme hAOX1 is characterized by a broad substrate specificity, oxidizing aromatic/aliphatic aldehydes into their corresponding carboxylic acids, and hydroxylating various heteroaromatic rings. The enzyme uses oxygen as terminal electron acceptor to produce hydrogen peroxide and superoxide during turnover. Since hAOX1 and, in particular, some natural variants produce not only H2O2 but also high amounts of superoxide, we investigated the effect of both ROS molecules on the enzymatic activity of hAOX1 in more detail. We compared hAOX1 to the high-O2 .--producing natural variant L438V for their time-dependent inactivation with H2O2/O2 .- during substrate turnover. We show that the inactivation of the hAOX1 wild-type enzyme is mainly based on the production of hydrogen peroxide, whereas for the variant L438V, both hydrogen peroxide and superoxide contribute to the time-dependent inactivation of the enzyme during turnover. Further, the level of inactivation was revealed to be substrate-dependent: using substrates with higher turnover numbers resulted in a faster inactivation of the enzymes. Analysis of the inactivation site of the enzyme identified a loss of the terminal sulfido ligand at the molybdenum active site by the produced ROS during turnover. SIGNIFICANCE STATEMENT: This work characterizes the substrate-dependent inactivation of human aldehyde oxidase 1 under turnover by reactive oxygen species and identifies the site of inactivation. The role of ROS in the inhibition of human aldehyde oxidase 1 will have a high impact on future studies.
Collapse
Affiliation(s)
- Claudia Garrido
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany
| | - Silke Leimkühler
- Institute of Biochemistry and Biology, Department of Molecular Enzymology, University of Potsdam, Potsdam, Germany
| |
Collapse
|
15
|
ROCK Inhibition as Potential Target for Treatment of Pulmonary Hypertension. Cells 2021; 10:cells10071648. [PMID: 34209333 PMCID: PMC8303917 DOI: 10.3390/cells10071648] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 02/07/2023] Open
Abstract
Pulmonary hypertension (PH) is a cardiovascular disease caused by extensive vascular remodeling in the lungs, which ultimately leads to death in consequence of right ventricle (RV) failure. While current drugs for PH therapy address the sustained vasoconstriction, no agent effectively targets vascular cell proliferation and tissue inflammation. Rho-associated protein kinases (ROCKs) emerged in the last few decades as promising targets for PH therapy, since ROCK inhibitors demonstrated significant anti-remodeling and anti-inflammatory effects. In this review, current aspects of ROCK inhibition therapy are discussed in relation to the treatment of PH and RV dysfunction, from cell biology to preclinical and clinical studies.
Collapse
|
16
|
Soltani S, Hallaj-Nezhadi S, Rashidi MR. A comprehensive review of in silico approaches for the prediction and modulation of aldehyde oxidase-mediated drug metabolism: The current features, challenges and future perspectives. Eur J Med Chem 2021; 222:113559. [PMID: 34119831 DOI: 10.1016/j.ejmech.2021.113559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 01/09/2023]
Abstract
The importance of aldehyde oxidase (AOX) in drug metabolism necessitates the development and application of the in silico rational drug design methods as an integral part of drug discovery projects for the early prediction and modulation of AOX-mediated metabolism. The current study represents an up-to-date and thorough review of in silico studies of AOX-mediated metabolism and modulation methods. In addition, the challenges and the knowledge gap that should be covered have been discussed. The importance of aldehyde oxidase (AOX) in drug metabolism is a hot topic in drug discovery. Different strategies are available for the modulation of the AOX-mediated metabolism of drugs. Application of the rational drug design methods as an integral part of drug discovery projects is necessary for the early prediction of AOX-mediated metabolism. The current study represents a comprehensive review of AOX molecular structure, AOX-mediated reactions, AOX substrates, AOX inhibition, approaches to modify AOX-mediated metabolism, prediction of AOX metabolism/substrates/inhibitors, and the AOX related structure-activity relationship (SAR) studies. Furthermore, an up-to-date and thorough review of in silico studies of AOX metabolism has been carried out. In addition, the challenges and the knowledge gap that should be covered in the scientific literature have been discussed in the current review.
Collapse
Affiliation(s)
- Somaieh Soltani
- Pharmaceutical Analysis Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Somayeh Hallaj-Nezhadi
- Drug Applied Research Center and Pharmacy Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Rashidi
- Stem Cell and Regenerative Medicine Institute and Pharmacy faculty, Tabriz University of Medical Sciences, Iran.
| |
Collapse
|
17
|
Basit A, Neradugomma NK, Wolford C, Fan PW, Murray B, Takahashi RH, Khojasteh SC, Smith BJ, Heyward S, Totah RA, Kelly EJ, Prasad B. Characterization of Differential Tissue Abundance of Major Non-CYP Enzymes in Human. Mol Pharm 2020; 17:4114-4124. [PMID: 32955894 DOI: 10.1021/acs.molpharmaceut.0c00559] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The availability of assays that predict the contribution of cytochrome P450 (CYP) metabolism allows for the design of new chemical entities (NCEs) with minimal oxidative metabolism. These NCEs are often substrates of non-CYP drug-metabolizing enzymes (DMEs), such as UDP-glucuronosyltransferases (UGTs), sulfotransferases (SULTs), carboxylesterases (CESs), and aldehyde oxidase (AO). Nearly 30% of clinically approved drugs are metabolized by non-CYP enzymes. However, knowledge about the differential hepatic versus extrahepatic abundance of non-CYP DMEs is limited. In this study, we detected and quantified the protein abundance of eighteen non-CYP DMEs (AO, CES1 and 2, ten UGTs, and five SULTs) across five different human tissues. AO was most abundantly expressed in the liver and to a lesser extent in the kidney; however, it was not detected in the intestine, heart, or lung. CESs were ubiquitously expressed with CES1 being predominant in the liver, while CES2 was enriched in the small intestine. Consistent with the literature, UGT1A4, UGT2B4, and UGT2B15 demonstrated liver-specific expression, whereas UGT1A10 expression was specific to the intestine. UGT1A1 and UGT1A3 were expressed in both the liver and intestine; UGT1A9 was expressed in the liver and kidney; and UGT2B17 levels were significantly higher in the intestine than in the liver. All five SULTs were detected in the liver and intestine, and SULT1A1 and 1A3 were detected in the lung. Kidney abundance was the most variable among the studied tissues, and overall, high interindividual variability (>15-fold) was observed for UGT2B17, CES2 (intestine), SULT1A1 (liver), UGT1A9, UGT2B7, and CES1 (kidney). These differential tissue abundance data can be integrated into physiologically based pharmacokinetic (PBPK) models for the prediction of non-CYP drug metabolism and toxicity in hepatic and extrahepatic tissues.
Collapse
Affiliation(s)
- Abdul Basit
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Naveen K Neradugomma
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Christopher Wolford
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Peter W Fan
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Bernard Murray
- Drug Metabolism and Pharmacokinetics Department, Gilead Sciences Inc., 324 Lakeside Drive, Foster City, California 94404, United States
| | - Ryan H Takahashi
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, MS 412a, South San Francisco, California 94080, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., 1 DNA Way, MS 412a, South San Francisco, California 94080, United States
| | - Bill J Smith
- Drug Metabolism and Pharmacokinetics Department, Gilead Sciences Inc., 324 Lakeside Drive, Foster City, California 94404, United States
| | - Scott Heyward
- BioIVT Inc., Baltimore, Maryland 21227, United States
| | - Rheem A Totah
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, United States
| | - Bhagwat Prasad
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| |
Collapse
|
18
|
Takahashi T, Luzum JA, Nicol MR, Jacobson PA. Pharmacogenomics of COVID-19 therapies. NPJ Genom Med 2020; 5:35. [PMID: 32864162 PMCID: PMC7435176 DOI: 10.1038/s41525-020-00143-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
A new global pandemic of coronavirus disease 2019 (COVID-19) has resulted in high mortality and morbidity. Currently numerous drugs are under expedited investigations without well-established safety or efficacy data. Pharmacogenomics may allow individualization of these drugs thereby improving efficacy and safety. In this review, we summarized the pharmacogenomic literature available for COVID-19 drug therapies including hydroxychloroquine, chloroquine, azithromycin, remdesivir, favipiravir, ribavirin, lopinavir/ritonavir, darunavir/cobicistat, interferon beta-1b, tocilizumab, ruxolitinib, baricitinib, and corticosteroids. We searched PubMed, reviewed the Pharmacogenomics Knowledgebase (PharmGKB®) website, Clinical Pharmacogenetics Implementation Consortium (CPIC) guidelines, the U.S. Food and Drug Administration (FDA) pharmacogenomics information in the product labeling, and the FDA pharmacogenomics association table. We found several drug-gene variant pairs that may alter the pharmacokinetics of hydroxychloroquine/chloroquine (CYP2C8, CYP2D6, SLCO1A2, and SLCO1B1); azithromycin (ABCB1); ribavirin (SLC29A1, SLC28A2, and SLC28A3); and lopinavir/ritonavir (SLCO1B1, ABCC2, CYP3A). We also identified other variants, that are associated with adverse effects, most notable in hydroxychloroquine/chloroquine (G6PD; hemolysis), ribavirin (ITPA; hemolysis), and interferon β -1b (IRF6; liver toxicity). We also describe the complexity of the risk for QT prolongation in this setting because of additive effects of combining more than one QT-prolonging drug (i.e., hydroxychloroquine/chloroquine and azithromycin), increased concentrations of the drugs due to genetic variants, along with the risk of also combining therapy with potent inhibitors. In conclusion, although direct evidence in COVID-19 patients is lacking, we identified potential actionable genetic markers in COVID-19 therapies. Clinical studies in COVID-19 patients are deemed warranted to assess potential roles of these markers.
Collapse
Affiliation(s)
- Takuto Takahashi
- Department of Experimental and Clinical Pharmacology, College of Pharmacy University of Minnesota, Minneapolis, MN USA
- Division of Hematology/Oncology/Blood and Marrow Transplantation, Department of Pediatrics, University of Minnesota, Minneapolis, MN USA
| | - Jasmine A. Luzum
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI USA
| | - Melanie R. Nicol
- Department of Experimental and Clinical Pharmacology, College of Pharmacy University of Minnesota, Minneapolis, MN USA
| | - Pamala A. Jacobson
- Department of Experimental and Clinical Pharmacology, College of Pharmacy University of Minnesota, Minneapolis, MN USA
| |
Collapse
|
19
|
Uehara S, Yoneda N, Higuchi Y, Yamazaki H, Suemizu H. Human Aldehyde Oxidase 1-Mediated Carbazeran Oxidation in Chimeric TK-NOG Mice Transplanted with Human Hepatocytes. Drug Metab Dispos 2020; 48:580-586. [PMID: 32357972 DOI: 10.1124/dmd.120.091090] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/10/2020] [Indexed: 02/13/2025] Open
Abstract
Carbazeran is a potent phosphodiesterase inhibitor with species-dependent metabolic profiles in rats, dogs, and humans. In this study, we investigated the aldehyde oxidase (AOX)-mediated oxidation of carbazeran to 4-oxo derivatives in chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene with humanized livers (humanized-liver mice). Liver cytosolic fractions from humanized-liver mouse effectively catalyzed carbazeran 4-oxidation with high affinity for the substrate, similar to those of the human liver cytosolic fractions and recombinant human AOX1 protein. Furthermore, hepatocytes prepared from humanized-liver mice and humans also exhibited substantial metabolism via carbazeran 4-oxidation. After a single oral administration of carbazeran (10 mg/kg), plasma levels of 4-oxo-carbazeran, N-desethyl-4-oxo-carbazeran, and 6,7-dimethoxy-1-[4-(hydroxy)-piperidino]-4-phthalazinone (three human metabolites formed via 4-oxidation) were higher in humanized-liver mice than in the control mice. In contrast, plasma levels of O-desmethylcarbazeran (a major metabolite in dogs) in control mice were higher than those in the humanized-liver mice. Relative excreted amounts of the three 4-oxidation-derived human-specific metabolites in the urine and feces were greater for humanized-liver mice than control mice, whereas the relative excreted amounts of O-desmethylcarbazeran were predominant in the urine and feces of control mice. Thus, the production of carbazeran 4-oxo derivatives was elevated in humanized-liver mice compared with control mice, in agreement with our in vitro enzyme-mediated oxidation data. These results suggest that hepatic human AOX1 functions in humanized-liver mice at the in vivo level and that humanized-liver mice may be useful for predicting drug metabolism in humans, at least with regard to human AOX1-dependent metabolism. SIGNIFICANCE STATEMENT: We found that the production of carbazeran 4-oxo derivatives was higher in humanized-liver mice than in control mice. These results were supported by the fact that carbazeran was rapidly metabolized to 4-oxo-carbazeran in humanized-liver mouse hepatocytes expressing human aldehyde oxidase 1. These results suggest that human aldehyde oxidase 1 is functional in humanized-liver mice in vivo and that chimeric NOD/Shi-scid IL2 receptor gamma-null mice expressing a herpes simplex virus type 1 thymidine kinase transgene transplanted with human hepatocytes may be a suitable model animal for predicting aldehyde oxidase-dependent biotransformation of drugs in humans.
Collapse
Affiliation(s)
- Shotaro Uehara
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan (S.U., N.Y., Y.H., H.S.) and the Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (H.Y.)
| | - Nao Yoneda
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan (S.U., N.Y., Y.H., H.S.) and the Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (H.Y.)
| | - Yuichiro Higuchi
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan (S.U., N.Y., Y.H., H.S.) and the Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (H.Y.)
| | - Hiroshi Yamazaki
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan (S.U., N.Y., Y.H., H.S.) and the Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (H.Y.)
| | - Hiroshi Suemizu
- Laboratory Animal Research Department, Central Institute for Experimental Animals, Kawasaki, Kanagawa, Japan (S.U., N.Y., Y.H., H.S.) and the Laboratory of Drug Metabolism and Pharmacokinetics, Showa Pharmaceutical University, Machida, Tokyo, Japan (H.Y.)
| |
Collapse
|
20
|
Formation and degradation of lipid droplets in human adipocytes and the expression of aldehyde oxidase (AOX). Cell Tissue Res 2019; 379:45-62. [DOI: 10.1007/s00441-019-03152-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
AbstractLipid droplet (LD) binding proteins in mammary glands and in adipocytes were previously compared and striking similar sets of these specific proteins demonstrated. Xanthine oxidoreductase (XOR) together with perilipins and the lactating mammary gland protein butyrophilin play an important role in the secretion process of LDs into milk ducts. In contrast, in adipose tissue and in adipocytes, mainly perilipins have been described. Moreover, XOR was reported in mouse adipose tissue and adipocyte culture cells as “novel regulator of adipogenesis”. This obvious coincidence of protein sets prompted us to revisit the formation of LDs in human-cultured adipocytes in more detail with special emphasis on the possibility of a LD association of XOR. We demonstrate by electron and immunoelectron microscopy new structural details on LD formation in adipocytes. Surprisingly, by immunological and proteomic analysis, we identify in contrast to previous data showing the enzyme XOR, predominantly the expression of aldehyde oxidase (AOX). AOX could be detected tightly linked to LDs when adipocytes were treated with starvation medium. In addition, the majority of cells show an enormous interconnected, tubulated mitochondria network. Here, we discuss that (1) XOR is involved—together with perilipins—in the secretion of LDs in alveolar epithelial cells of the lactating mammary gland and is important in the transcytosis pathway of capillary endothelial cells. (2) In cells, where LDs are not secreted, XOR cannot be detected at the protein level, whereas in contrast in these cases, AOX is often present. We detect AOX in adipocytes together with perilipins and find evidence that these proteins might direct LDs to mitochondria. Finally, we here report for the first time the exclusive and complementary localization of XOR and AOX in diverse cell types.
Collapse
|