1
|
Vasilogianni AM, Achour B, Al-Majdoub ZM, Peters SA, Barber J, Rostami-Hodjegan A. The quest to define cancer-specific systems parameters for personalized dosing in oncology. Expert Opin Drug Metab Toxicol 2025:1-17. [PMID: 40042382 DOI: 10.1080/17425255.2025.2476560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Clinical trials in oncology initially recruit heterogeneous populations, without catering for all types of variability. The target cohort is often not representative, leading to variability in pharmacokinetics (PK). To address enrollment challenges in clinical trials, physiologically based pharmacokinetic models (PBPK) models can be used as a guide in the absence of large clinical studies. These models require patient-specific systems data relevant to the handling of drugs in the body for each type of cancer, which are scarce. AREAS COVERED This review explores system parameters affecting PK in cancer and highlights important gaps in data. Changes in drug-metabolizing enzymes (DMEs) and transporters have not been fully investigated in cancer. Their impaired expression can significantly affect capacity for drug elimination. Finally, the use of PBPK modeling for precision dosing in oncology is highlighted. Google Scholar and PubMed were mainly used for literature search, without date restriction. EXPERT OPINION Model-informed precision dosing is useful for dosing in sub-groups of cancer patients, which might not have been included in clinical trials. Systems parameters are not fully characterized in cancer cohorts, which are required in PBPK models. Generation of such data and application of cancer models in clinical practice should be encouraged.
Collapse
Affiliation(s)
- Areti-Maria Vasilogianni
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Darmstadt, Germany
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co., Ingelheim am Rhein, Germany
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- Certara Predictive Technologies (CPT), Simcyp Division, Sheffield, UK
| |
Collapse
|
2
|
Xie Y, Fan Y, Liu X, Li Z, Liu S. 4D-DIA-based proteomics analysis reveals the protective effects of Pidanjiangtang granules in IGT rat model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119012. [PMID: 39481621 DOI: 10.1016/j.jep.2024.119012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Pidanjiangtang (PDJT) formula was founded on the "Pidan" theory from the "Nei Jing." PDJT is considered to eliminate the accumulation of pathological products, remove heat sources, and prevent damage to organs such as the liver and islets. It is widely used in clinical practice to treat impaired glucose tolerance (IGT). However, the bioactive ingredients and underlying mechanisms are still unclear and need further investigation. OBJECTIVE This study aimed to determine the therapeutic effect of PDJT on IGT rats and explore the mechanism of PDJT intervention on IGT by four-dimensional independent data acquisition (4D-DIA) proteomics analysis. MATERIALS AND METHODS The IGT model was established by a high-fat diet combined with Streptozotocin (STZ) injection. The IGT rats were treated with low, medium, and high doses of PDJT orally for 42 days and compared with the Metformin positive control group. The therapeutic effects of PDJT on IGT rats were evaluated using the oral glucose tolerance test (OGTT), serum lipoprotein detection, insulin detection, liver histopathology, and hepatic steatosis assessment. 4D-DIA proteomics analysis was used to explore the differential proteins (DEPs) and potential pathways of PDJT. Finally, Western blotting and ELISA techniques were used to verify DEPs and major targets. RESULTS PDJT can enhance glucose metabolism, restore islet β cell function, regulate lipoprotein metabolism, reduce hepatic steatosis, and consequently slow down the progression of IGT. In the proteomic analysis, a total of 355 DEPs were identified, and critical proteins were validated. The results indicated that the JAK2/STAT1 signaling pathway plays a pivotal role in the effects of PDJT. IκB-ζ may be a potential target for PDJT in regulating the inflammatory response of IGT. CONCLUSION PDJT is an effective formula for improving IGT, with its potential mechanism linked to the JAK2/STAT1/IκB-ζ signaling pathway. This study offers a novel approach to investigating the mechanisms of TCM formula through proteomics and offers new insight into exploring TCM treatment for IGT.
Collapse
Affiliation(s)
- Yu Xie
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yue Fan
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Liu
- Beijing University of Chinese Medicine, Beijing, China; Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zirong Li
- Beijing University of Chinese Medicine, Beijing, China
| | - Shangjian Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
3
|
Prasad B, Al-Majdoub ZM, Wegler C, Rostami-Hodjegan A, Achour B. Quantitative Proteomics for Translational Pharmacology and Precision Medicine: State of The Art and Future Outlook. Drug Metab Dispos 2024; 52:1208-1216. [PMID: 38821856 DOI: 10.1124/dmd.124.001600] [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: 02/28/2024] [Revised: 05/02/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024] Open
Abstract
Over the past 20 years, quantitative proteomics has contributed a wealth of protein expression data, which are currently used for a variety of systems pharmacology applications, as a complement or a surrogate for activity of the corresponding proteins. A symposium at the 25th North American International Society for the Study of Xenobiotics meeting, in Boston, in September 2023, was held to explore current and emerging applications of quantitative proteomics in translational pharmacology and strategies for improved integration into model-informed drug development based on practical experience of each of the presenters. A summary of the talks and discussions is presented in this perspective alongside future outlook that was outlined for future meetings. SIGNIFICANCE STATEMENT: This perspective explores current and emerging applications of quantitative proteomics in translational pharmacology and precision medicine and outlines the outlook for improved integration into model-informed drug development.
Collapse
Affiliation(s)
- Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Zubida M Al-Majdoub
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Christine Wegler
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Amin Rostami-Hodjegan
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Brahim Achour
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (B.P.); Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (Z.M.A.-M., A.R.-H.); Department of Plant Physiology, Umeå University, Umeå, Sweden (C.W.); Certara UK, Sheffield, United Kingdom (A.R.-H.); and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| |
Collapse
|
4
|
Wang Z, Liu PK, Li L. A Tutorial Review of Labeling Methods in Mass Spectrometry-Based Quantitative Proteomics. ACS MEASUREMENT SCIENCE AU 2024; 4:315-337. [PMID: 39184361 PMCID: PMC11342459 DOI: 10.1021/acsmeasuresciau.4c00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 08/27/2024]
Abstract
Recent advancements in mass spectrometry (MS) have revolutionized quantitative proteomics, with multiplex isotope labeling emerging as a key strategy for enhancing accuracy, precision, and throughput. This tutorial review offers a comprehensive overview of multiplex isotope labeling techniques, including precursor-based, mass defect-based, reporter ion-based, and hybrid labeling methods. It details their fundamental principles, advantages, and inherent limitations along with strategies to mitigate the limitation of ratio-distortion. This review will also cover the applications and latest progress in these labeling techniques across various domains, including cancer biomarker discovery, neuroproteomics, post-translational modification analysis, cross-linking MS, and single-cell proteomics. This Review aims to provide guidance for researchers on selecting appropriate methods for their specific goals while also highlighting the potential future directions in this rapidly evolving field.
Collapse
Affiliation(s)
- Zicong Wang
- School
of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| | - Peng-Kai Liu
- Biophysics
Graduate program, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| | - Lingjun Li
- School
of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
- Biophysics
Graduate program, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
- Department
of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
- Lachman
Institute for Pharmaceutical Development, School of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
- Wisconsin
Center for NanoBioSystems, School of Pharmacy, University of Wisconsin—Madison, Madison, Wisconsin 53705, United States
| |
Collapse
|
5
|
Wang P, Shen Y, Manaenko A, Liu F, Yang W, Xiao Z, Li P, Ran Y, Dang R, He Y, Wu Q, Xie P, Li Q. TMT-based quantitative proteomics reveals the protective mechanism of tenuigenin after experimental intracerebral hemorrhage in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117213. [PMID: 37739103 DOI: 10.1016/j.jep.2023.117213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 09/17/2023] [Accepted: 09/20/2023] [Indexed: 09/24/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tenuigenin (TNG) is an extract obtained from Polygalae Radix. It possesses anti-inflammatory, antioxidant, and neuroprotective properties. However, the potential mechanism of TNG in intracerebral hemorrhage (ICH) has not been well studied. AIM OF THE STUDY In the present study, we aimed to identify the prospective mechanism of TNG in treating ICH. MATERIALS AND METHODS A total of 120 mice were divided into five groups: Sham group, ICH + vehicle group, ICH + TNG(8 mg/kg), ICH + TNG(16 mg/kg), and ICH + TNG(32 mg/kg). The modified Garcia test and beam walking test were carried out at 24 h and 72 h after ICH. Brain water content, haematoma volume and hemoglobin content examinations were performed at 72 h after ICH. TMT-based quantitative proteomics combined with bioinformatics analysis methods was used to distinguish differentially expressed proteins (DEPs) to explore potential pharmacological mechanisms. Western blotting was performed to validate representative proteins. RESULTS Our results showed that the optimal dose of TNG was 16 mg/kg, which could markedly improve neurological functions, and reduce cerebral oedema, haematoma volume and hemoglobin levels 72 h after ICH. A total of 404 DEPs (353 up-and 51 downregulated) were identified in the ICH + vehicle vs. sham group, while 342 DEPs (306 up-and 36 downregulated) and 76 DEPs (28 up-and 48 downregulated) were quantified in the TNG vs. sham group and TNG vs. ICH + vehicle group, respectively. In addition, a total of 26 DEPs were selected according to strict criteria. Complement and coagulation cascades were the most significantly enriched pathways, and two proteins (MBL-C and Car1) were further validated as hub molecules. CONCLUSIONS Our results suggested that the therapeutic effects of TNG on ICH were closely associated with the complement system, and that MBL-C and Car1 might be potential targets of TNG for the treatment of ICH.
Collapse
Affiliation(s)
- Peng Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - YiQing Shen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Anatol Manaenko
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - FangYu Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - WenSong Yang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - ZhongSong Xiao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - PeiZheng Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - YuXin Ran
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - RuoZhi Dang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong He
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - QingYuan Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Qi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China; NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
6
|
Goelen J, Farrell G, McGeehan J, Titman CM, J W Rattray N, Johnson TN, Horniblow RD, Batchelor HK. Quantification of drug metabolising enzymes and transporter proteins in the paediatric duodenum via LC-MS/MS proteomics using a QconCAT technique. Eur J Pharm Biopharm 2023; 191:68-77. [PMID: 37625656 DOI: 10.1016/j.ejpb.2023.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
Characterising the small intestine absorptive membrane is essential to enable prediction of the systemic exposure of oral formulations. In particular, the ontogeny of key intestinal Drug Metabolising Enzymes and Transporter (DMET) proteins involved in drug disposition needs to be elucidated to allow for accurate prediction of the PK profile of drugs in the paediatric cohort. Using pinch biopsies from the paediatric duodenum (n = 36; aged 11 months to 15 years), the abundance of 21 DMET proteins and two enterocyte markers were quantified via LC-MS/MS. An established LCMS nanoflow method was translated to enable analysis on a microflow LC system, and a new stable-isotope-labelled QconCAT standard developed to enable quantification of these proteins. Villin-1 was used to standardise abundancy values. The observed abundancies and ontogeny profiles, agreed with adult LC-MS/MS-based data, and historic paediatric data obtained via western blotting. A linear trend with age was observed for duodenal CYP3A4 and CES2 only. As this work quantified peptides on a pinch biopsy coupled with a microflow method, future studies using a wider population range are very feasible. Furthermore, this DMET ontogeny data can be used to inform paediatric PBPK modelling and to enhance the understanding of oral drug absorption and gut bioavailability in paediatric populations.
Collapse
Affiliation(s)
- Jan Goelen
- School of Pharmacy, University of Birmingham, Birmingham B15 2TT, UK; Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | - Gillian Farrell
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | | | | | - Nicholas J W Rattray
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK
| | | | - Richard D Horniblow
- School of Biomedical Science, University of Birmingham, Birmingham B15 2TT, UK
| | - Hannah K Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
7
|
Bai JPF, Yu LR. Modeling Clinical Phenotype Variability: Consideration of Genomic Variations, Computational Methods, and Quantitative Proteomics. J Pharm Sci 2023; 112:904-908. [PMID: 36279954 DOI: 10.1016/j.xphs.2022.10.016] [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: 09/07/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
Advances in biomedical and computer technologies have presented the modeling community the opportunity for mechanistically modeling and simulating the variability in a disease phenotype or in a drug response. The capability to quantify response variability can inform a drug development program. Quantitative systems pharmacology scientists have published various computational approaches for creating virtual patient populations (VPops) to model and simulate drug response variability. Genomic variations can impact disease characteristics and drug exposure and response. Quantitative proteomics technologies are increasingly used to facilitate drug discovery and development and inform patient care. Incorporating variations in genomics and quantitative proteomics may potentially inform creation of VPops to model and simulate virtual patient trials, and may help account for, in a predictive manner, phenotypic variations observed clinically.
Collapse
Affiliation(s)
- Jane P F Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD 20903, USA.
| | - Li-Rong Yu
- Division of Systems Biology, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR 72079, USA
| |
Collapse
|
8
|
Vasilogianni AM, Al-Majdoub ZM, Achour B, Peters SA, Rostami-Hodjegan A, Barber J. Proteomic quantification of receptor tyrosine kinases involved in the development and progression of colorectal cancer liver metastasis. Front Oncol 2023; 13:1010563. [PMID: 36890818 PMCID: PMC9986493 DOI: 10.3389/fonc.2023.1010563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Introduction Alterations in expression and activity of human receptor tyrosine kinases (RTKs) are associated with cancer progression and in response to therapeutic intervention. Methods Thus, protein abundance of 21 RTKs was assessed in 15 healthy and 18 cancerous liver samples [2 primary and 16 colorectal cancer liver metastasis (CRLM)] matched with non-tumorous (histologically normal) tissue, by a validated QconCAT-based targeted proteomic approach. Results It was demonstrated, for the first time, that the abundance of EGFR, INSR, VGFR3 and AXL, is lower in tumours relative to livers from healthy individuals whilst the opposite is true for IGF1R. EPHA2 was upregulated in tumour compared with histologically normal tissue surrounding it. PGFRB levels were higher in tumours relative to both histologically normal tissue surrounding tumour and tissues taken from healthy individuals. The abundances of VGFR1/2, PGFRA, KIT, CSF1R, FLT3, FGFR1/3, ERBB2, NTRK2, TIE2, RET, and MET were, however, comparable in all samples. Statistically significant, but moderate correlations were observed (Rs > 0.50, p < 0.05) for EGFR with INSR and KIT. FGFR2 correlated with PGFRA and VGFR1 with NTRK2 in healthy livers. In non-tumorous (histologically normal) tissues from cancer patients, there were correlations between TIE2 and FGFR1, EPHA2 and VGFR3, FGFR3 and PGFRA (p < 0.05). EGFR correlated with INSR, ERBB2, KIT and EGFR, and KIT with AXL and FGFR2. In tumours, CSF1R correlated with AXL, EPHA2 with PGFRA, and NTRK2 with PGFRB and AXL. Sex, liver lobe and body mass index of donors had no impact on the abundance of RTKs, although donor age showed some correlations. RET was the most abundant of these kinases in non-tumorous tissues (~35%), while PGFRB was the most abundant RTK in tumours (~47%). Several correlations were also observed between the abundance of RTKs and proteins relevant to drug pharmacokinetics (enzymes and transporters). Discussion DiscussionThis study quantified perturbation to the abundance of several RTKs in cancer and the value generated in this study can be used as input to systems biology models defining liver cancer metastases and biomarkers of its progression.
Collapse
Affiliation(s)
- Areti-Maria Vasilogianni
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, United Kingdom.,Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Darmstadt, Germany.,Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co., KG, Ingelheim am Rhein, Germany
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, United Kingdom.,Simcyp Division, Certara Inc., Sheffield, United Kingdom
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
9
|
Alrubia S, Al-Majdoub ZM, Achour B, Rostami-Hodjegan A, Barber J. Quantitative Assessment of the Impact of Crohn's Disease on Protein Abundance of Human Intestinal Drug-Metabolising Enzymes and Transporters. J Pharm Sci 2022; 111:2917-2929. [PMID: 35872023 DOI: 10.1016/j.xphs.2022.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/17/2022] [Accepted: 07/17/2022] [Indexed: 10/17/2022]
Abstract
Crohn's disease affects the mucosal layer of the intestine, predominantly ileum and colon segments, with the potential to affect the expression of intestinal enzymes and transporters, and consequently, oral drug bioavailability. We carried out a quantitative proteomic analysis of inflamed and non-inflamed ileum and colon tissues from Crohn's disease patients and healthy donors. Homogenates from samples in each group were pooled and protein abundance determined by liquid chromatography-mass spectrometry (LC-MS). In inflamed Crohn's ileum, CYP3A4, CYP20A1, CYP51A1, ADH1B, ALPI, FOM1, SULT1A2, SULT1B1 and ABCB7 showed ≥10-fold reduction in abundance compared with healthy baseline. By contrast, only MGST1 showed ≥10 fold reduction in inflamed colon. Ileal UGT1A1, MGST1, MGST2, and MAOA levels increased by ≥2 fold in Crohn's patients, while only ALPI showed ≥2 fold increase in the colon. Counter-intuitively, non-inflamed ileum had a higher magnitude of fold change than inflamed tissue when compared with healthy tissue. Marked but non-uniform alterations were observed in the expression of various enzymes and transporters in ileum and colon compared with healthy samples. Modelling will allow improved understanding of the variable effects of Crohn's disease on bioavailability of orally administered drugs.
Collapse
Affiliation(s)
- Sarah Alrubia
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK; Pharmaceutical Chemistry Department, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK; Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island, USA
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK; Certara UK Ltd, Simcyp Division, Level 2-Acero, 1 Concourse Way, Sheffield, UK
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
10
|
Insights into the use of genetically modified decellularized biomaterials for tissue engineering and regenerative medicine. Adv Drug Deliv Rev 2022; 188:114413. [PMID: 35777666 DOI: 10.1016/j.addr.2022.114413] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/25/2022] [Accepted: 06/25/2022] [Indexed: 11/24/2022]
Abstract
Various modifications have been performed on biomaterials to improve their applications in tissue engineering and regenerative medicine. However, the challenges of immunogenicity and biocompatibility existed since the application of biomaterials. As a method to solve this problem, the decellularization process removes most living cells from biomaterials to minimize their immunogenicity; and preserves the native structures and compositions that favour cell growth and the subsequent construction of functional tissue. On the other hand, genetic modification of biomaterials aims to achieve specific functions (low immunogenicity, osteogenesis, etc.) or analyse the genetic mechanisms underlying some diseases (cardiac dysfunction, liver fibrosis, etc.). The combination of decellularization and gene modification is highly superior to biomaterials; thus, we must obtain a deeper understanding of these novel biomaterials. In this review, we summarize the fabrication approaches and current applications of genetically modified decellularized biomaterials and then discuss their disadvantages and corresponding future perspectives.
Collapse
|
11
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
12
|
Barber J, Al-Majdoub ZM, Couto N, Vasilogianni AM, Tillmann A, Alrubia S, Rostami-Hodjegan A, Achour B. Label-Free but Still Constrained: Assessment of Global Proteomic Strategies for the Quantification of Hepatic Enzymes and Transporters. Drug Metab Dispos 2022; 50:762-769. [PMID: 35307650 DOI: 10.1124/dmd.121.000780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 03/04/2022] [Indexed: 02/13/2025] Open
Abstract
Building and refining pharmacology models require "system" data derived from tissues and in vitro systems analyzed by quantitative proteomics. Label-free global proteomics offers a wide scope of analysis, allowing simultaneous quantification of thousands of proteins per sample. The data generated from such analysis offer comprehensive protein expression profiles that can address existing gaps in models. In this study, we assessed the performance of three widely used label-free proteomic methods, "high N" ion intensity approach (HiN), intensity-based absolute quantification (iBAQ) and total protein approach (TPA), in relation to the quantification of enzymes and transporters in 27 human liver microsomal samples. Global correlations between the three methods were highly significant (R2 > 0.70, P < 0.001, n = 2232 proteins). Absolute abundances of 57 pharmacokinetic targets measured by standard-based label-free methods (HiN and iBAQ) showed good agreement, whereas the TPA overestimated abundances by two- to threefold. Relative abundance distribution of enzymes was similar for the three methods, while differences were observed with TPA in the case of transporters. Variability (CV) was similar across methods, with consistent between-sample relative quantification. The back-calculated amount of protein in the samples based on each method was compared with the nominal protein amount analyzed in the proteomic workflow, revealing overall agreement with data from the HiN method with bovine serum albumin as standard. The findings herein present a critique of label-free proteomic data relevant to pharmacokinetics and evaluate the possibility of retrospective analysis of historic datasets. SIGNIFICANCE STATEMENT: This study provides useful insights for using label-free methods to generate abundance data applicable for populating pharmacokinetic models. The data demonstrated overall correlation between intensity-based label-free proteomic methods (HiN, iBAQ and TPA), whereas iBAQ and TPA overestimated the total amount of protein in the samples. The extent of overestimation can provide a means of normalization to support absolute quantification. Importantly, between-sample relative quantification was consistent (similar variability) across methods.
Collapse
Affiliation(s)
- Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Narciso Couto
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Areti-Maria Vasilogianni
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Annika Tillmann
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Sarah Alrubia
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester, United Kingdom (J.B., Z.M.A.-M., N.C., A.-M.V., A.T., S.A., A.R.-H., B.A.) Simcyp Division, Certara, Sheffield, United Kingdom (A.R.-H.) and Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, the University of Rhode Island, Kingston, Rhode Island (B.A.)
| |
Collapse
|
13
|
Enhanced anticancer activities of curcumin-loaded green gum acacia-based silver nanoparticles against melanoma and breast cancer cells. APPLIED NANOSCIENCE 2021. [DOI: 10.1007/s13204-021-02176-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
14
|
Claridge B, Lozano J, Poh QH, Greening DW. Development of Extracellular Vesicle Therapeutics: Challenges, Considerations, and Opportunities. Front Cell Dev Biol 2021; 9:734720. [PMID: 34616741 PMCID: PMC8488228 DOI: 10.3389/fcell.2021.734720] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 07/30/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) hold great promise as therapeutic modalities due to their endogenous characteristics, however, further bioengineering refinement is required to address clinical and commercial limitations. Clinical applications of EV-based therapeutics are being trialed in immunomodulation, tissue regeneration and recovery, and as delivery vectors for combination therapies. Native/biological EVs possess diverse endogenous properties that offer stability and facilitate crossing of biological barriers for delivery of molecular cargo to cells, acting as a form of intercellular communication to regulate function and phenotype. Moreover, EVs are important components of paracrine signaling in stem/progenitor cell-based therapies, are employed as standalone therapies, and can be used as a drug delivery system. Despite remarkable utility of native/biological EVs, they can be improved using bio/engineering approaches to further therapeutic potential. EVs can be engineered to harbor specific pharmaceutical content, enhance their stability, and modify surface epitopes for improved tropism and targeting to cells and tissues in vivo. Limitations currently challenging the full realization of their therapeutic utility include scalability and standardization of generation, molecular characterization for design and regulation, therapeutic potency assessment, and targeted delivery. The fields' utilization of advanced technologies (imaging, quantitative analyses, multi-omics, labeling/live-cell reporters), and utility of biocompatible natural sources for producing EVs (plants, bacteria, milk) will play an important role in overcoming these limitations. Advancements in EV engineering methodologies and design will facilitate the development of EV-based therapeutics, revolutionizing the current pharmaceutical landscape.
Collapse
Affiliation(s)
- Bethany Claridge
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Jonathan Lozano
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - David W. Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
15
|
El-Khateeb E, Al-Majdoub ZM, Rostami-Hodjegan A, Barber J, Achour B. Proteomic Quantification of Changes in Abundance of Drug-Metabolizing Enzymes and Drug Transporters in Human Liver Cirrhosis: Different Methods, Similar Outcomes. Drug Metab Dispos 2021; 49:610-618. [PMID: 34045218 DOI: 10.1124/dmd.121.000484] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Model-based assessment of the effects of liver disease on drug pharmacokinetics requires quantification of changes in enzymes and transporters responsible for drug metabolism and disposition. Different proteomic methods are currently used for protein quantification in tissues and in vitro systems, each with specific procedures and requirements. The outcome of quantitative proteomic assays using four different methods (one targeted and three label-free) applied to the same sample set was compared in this study. Three pooled cirrhotic liver microsomal samples corresponding to cirrhosis with nonalcoholic fatty liver disease, biliary disease, or cancer and a control microsomal pool were analyzed using quantification concatemer-based targeted proteomics, the total protein approach (TPA), high three ion intensity (Hi3) approach, and intensity-based absolute quantification (iBAQ) to determine the absolute and relative abundance in disease compared with control. The relative abundance data provided a "disease perturbation factor" (DPF) for each target protein. Absolute and relative abundances generated by standard-based label-free methods (iBAQ and Hi3) showed good agreement with targeted proteomics (limited bias and scatter), but TPA (standard-free method) overestimated absolute abundances by approximately 2-fold. The DPF was consistent between different proteomic methods but varied between enzymes and transporters, indicating discordance of effects of cirrhosis on various metabolism-related proteins. The DPF ranged from no change (e.g., for glucuronosyltransferase-1A6 in nonalcoholic fatty liver disease group) to less than 0.3 (e.g., carboxylesterases-1 in cirrhosis of biliary origin). SIGNIFICANCE STATEMENT: This study demonstrated that relative changes in enzymes and transporters (DPF) are independent of the quantitative proteomic methods used. Standard-based label-free methods, such as high three ion intensity (Hi3) and intensity-based absolute quantification (iBAQ) methods, were less biased and more precise than the total protein approach (TPA) when compared with targeted data. The DPF reconciled differences across proteomic methods observed with absolute levels. Using this approach, differences were revealed in the expression of enzymes/transporters in cirrhosis associated with different etiologies.
Collapse
Affiliation(s)
- Eman El-Khateeb
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK (E.E.-K., Z.M.A.-M., A.R.-H., J.B., B.A.); Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt (E.E.-K.); and Certara UK Ltd. (Simcyp Division), Sheffield, UK (A.R.-H.)
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK (E.E.-K., Z.M.A.-M., A.R.-H., J.B., B.A.); Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt (E.E.-K.); and Certara UK Ltd. (Simcyp Division), Sheffield, UK (A.R.-H.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK (E.E.-K., Z.M.A.-M., A.R.-H., J.B., B.A.); Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt (E.E.-K.); and Certara UK Ltd. (Simcyp Division), Sheffield, UK (A.R.-H.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK (E.E.-K., Z.M.A.-M., A.R.-H., J.B., B.A.); Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt (E.E.-K.); and Certara UK Ltd. (Simcyp Division), Sheffield, UK (A.R.-H.)
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK (E.E.-K., Z.M.A.-M., A.R.-H., J.B., B.A.); Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt (E.E.-K.); and Certara UK Ltd. (Simcyp Division), Sheffield, UK (A.R.-H.)
| |
Collapse
|
16
|
El-Khateeb E, Darwich AS, Achour B, Athwal V, Rostami-Hodjegan A. Review article: time to revisit Child-Pugh score as the basis for predicting drug clearance in hepatic impairment. Aliment Pharmacol Ther 2021; 54:388-401. [PMID: 34218453 DOI: 10.1111/apt.16489] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/14/2021] [Accepted: 06/04/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prescription information for many drugs entering the market lacks dosage guidance for hepatic impairment. Dedicated studies for assessing the fate of drugs in hepatic impairment commonly stratify patients using Child-Pugh score. Child-Pugh is a prognostic clinical score with limitations in reflecting the liver's metabolic capacity. AIMS To demonstrate the need for better drug dosing approaches in hepatic impairment, summarise the current status, identify knowledge gaps related to drug kinetic parameters in hepatic impairment, propose solutions for predicting the liver disease impact on drug exposure and discuss barriers to dosing guidance in those patients. METHODS Relevant reports on dosage adjustment in hepatic impairment were analysed concerning the prediction of the impairment impact on drug kinetics using physiologically-based pharmacokinetic (PBPK) modelling. RESULTS PBPK models are suggested as a potential framework to understand drug clearance changes in hepatic impairment. Quantifying changes in abundance and activity of drug-metabolising enzymes and transporters, understanding the impact of shunting, and accounting for interindividual variations in drug absorption could help in extending the success of these models in hepatically-impaired populations. These variables might not correlate with Child-Pugh score as a whole. Therefore, new metabolic activity markers, imaging techniques and other scoring systems are proposed to either support or substitute Child-Pugh score. CONCLUSIONS Many physiological changes in hepatic impairment determining the fate of drugs do not necessarily correlate with Child-Pugh score. Quantifying these changes in individual patients is essential in future hepatic impairment studies. Further studies assessing Child-Pugh alternatives are recommended to allow better prediction of drug exposure.
Collapse
Affiliation(s)
- Eman El-Khateeb
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Adam S Darwich
- Logistics and Informatics in Health Care, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, Stockholm, Sweden
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Varinder Athwal
- Wellcome Centre for Cell-Matrix Research, Division of Diabetes, Endocrinology and Gastroenterology, University of Manchester, Manchester, UK.,Research and Innovation Division, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK.,Certara UK Ltd. (Simcyp Division), Sheffield, UK
| |
Collapse
|
17
|
Couto N, Newton JRA, Russo C, Karunakaran E, Achour B, Al-Majdoub ZM, Sidaway J, Rostami-Hodjegan A, Clench MR, Barber J. Label-Free Quantitative Proteomics and Substrate-Based Mass Spectrometry Imaging of Xenobiotic Metabolizing Enzymes in Ex Vivo Human Skin and a Human Living Skin Equivalent Model. Drug Metab Dispos 2021; 49:39-52. [PMID: 33139459 DOI: 10.1124/dmd.120.000168] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/08/2020] [Indexed: 01/15/2023] Open
Abstract
We report for the first time label-free quantification of xenobiotic metabolizing enzymes (XME), transporters, redox enzymes, proteases, and nucleases in six human skin explants and a three-dimensional living skin equivalent model from LabSkin. We aimed to evaluate the suitability of LabSkin as an alternative to animal testing for the development of topical formulations. More than 2000 proteins were identified and quantified from total cellular protein. Alcohol dehydrogenase 1C, the most abundant phase I XME in human skin, and glutathione S-transferase pi 1, the most abundant phase II XME in human skin, were present in similar abundance in LabSkin. Several esterases were quantified and esterase activity was confirmed in LabSkin using substrate-based mass spectrometry imaging. No cytochrome P450 (P450) activity was observed for the substrates tested, in agreement with the proteomics data, where the cognate P450s were absent in both human skin and LabSkin. Label-free protein quantification allowed insights into other related processes such as redox homeostasis and proteolysis. For example, the most abundant antioxidant enzymes were thioredoxin and peroxiredoxin-1. This systematic determination of functional equivalence between human skin and LabSkin is a key step toward the construction of a representative human in vitro skin model, which can be used as an alternative to current animal-based tests for chemical safety and for predicting dosage of topically administered drugs. SIGNIFICANCE STATEMENT: The use of label-free quantitative mass spectrometry to elucidate the abundance of xenobiotic metabolizing enzymes, transporters, redox enzymes, proteases, and nucleases in human skin enhance our understanding of the skin physiology and biotransformation of topical drugs and cosmetics. This will help to develop mathematical models to predict drug metabolism in human skin and to develop more robust in vitro engineered human skin tissue as alternatives to animal testing.
Collapse
Affiliation(s)
- Narciso Couto
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Jillian R A Newton
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Cristina Russo
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Esther Karunakaran
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Brahim Achour
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Zubida M Al-Majdoub
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - James Sidaway
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Amin Rostami-Hodjegan
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Malcolm R Clench
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| | - Jill Barber
- Department of Chemical and Biological Engineering (N.C., E.K.) and Sheffield Collaboratorium for Antimicrobial Resistance and Biofilms (SCARAB) (N.C., E.K.), University of Sheffield, Sheffield, United Kingdom; Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., B.A., Z.M.A.-M., A.R.-H., J.B.); Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom (J.R.A.N., C.R., M.R.C.); Phenotox Ltd., Bollington, United Kingdom (J.S.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (A.R.-H.)
| |
Collapse
|
18
|
Al‐Majdoub ZM, Achour B, Couto N, Howard M, Elmorsi Y, Scotcher D, Alrubia S, El‐Khateeb E, Vasilogianni A, Alohali N, Neuhoff S, Schmitt L, Rostami‐Hodjegan A, Barber J. Mass spectrometry-based abundance atlas of ABC transporters in human liver, gut, kidney, brain and skin. FEBS Lett 2020; 594:4134-4150. [PMID: 33128234 PMCID: PMC7756589 DOI: 10.1002/1873-3468.13982] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/24/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
ABC transporters (ATP-binding cassette transporter) traffic drugs and their metabolites across membranes, making ABC transporter expression levels a key factor regulating local drug concentrations in different tissues and individuals. Yet, quantification of ABC transporters remains challenging because they are large and low-abundance transmembrane proteins. Here, we analysed 200 samples of crude and membrane-enriched fractions from human liver, kidney, intestine, brain microvessels and skin, by label-free quantitative mass spectrometry. We identified 32 (out of 48) ABC transporters: ABCD3 was the most abundant in liver, whereas ABCA8, ABCB2/TAP1 and ABCE1 were detected in all tissues. Interestingly, this atlas unveiled that ABCB2/TAP1 may have TAP2-independent functions in the brain and that biliary atresia (BA) and control livers have quite different ABC transporter profiles. We propose that meaningful biological information can be derived from a direct comparison of these data sets.
Collapse
Affiliation(s)
- Zubida M. Al‐Majdoub
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Narciso Couto
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Martyn Howard
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Yasmine Elmorsi
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| | - Sarah Alrubia
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Chemistry DepartmentCollege of PharmacyKing Saud UniversityRiyadhSaudi Arabia
| | - Eman El‐Khateeb
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Clinical Pharmacy DepartmentFaculty of PharmacyTanta UniversityEgypt
| | | | - Noura Alohali
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Pharmaceutical Practice DepartmentCollege of PharmacyPrincess Noura Bint Abdul Rahman UniversityRiyadhSaudi Arabia
| | | | - Lutz Schmitt
- Institute of BiochemistryHeinrich Heine University DüsseldorfGermany
| | - Amin Rostami‐Hodjegan
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
- Simcyp DivisionCertara UK LtdSheffieldUK
| | - Jill Barber
- Centre for Applied Pharmacokinetic ResearchSchool of Health SciencesUniversity of ManchesterUK
| |
Collapse
|
19
|
An Investigation into Proteomic Constituents of Cerebrospinal Fluid in Patients with Chronic Peripheral Neuropathic Pain Medicated with Opioids- a Pilot Study. J Neuroimmune Pharmacol 2020; 16:634-650. [PMID: 33219474 DOI: 10.1007/s11481-020-09970-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/30/2020] [Indexed: 12/25/2022]
Abstract
The pharmacodynamics of opioids for chronic peripheral neuropathic pain are complex and likely extend beyond classical opioid receptor theory. Preclinical evidence of opioid modulation of central immune signalling has not been identified in vivo in humans. Examining the cerebrospinal fluid (CSF) of patients medicated with opioids is required to identify potential pharmacodynamic mechanisms. We compared CSF samples of chronic peripheral neuropathic pain patients receiving opioids (n = 7) versus chronic peripheral neuropathic pain patients not taking opioids (control group, n = 13). Baseline pain scores with demographics were recorded. Proteome analysis was performed using mass spectrometry and secreted neuropeptides were measured by enzyme-linked immunosorbent assay. Based on Gene Ontology analysis, proteins involved in the positive regulation of nervous system development and myeloid leukocyte activation were increased in patients taking opioids versus the control group. The largest decrease in protein expression in patients taking opioids were related to neutrophil mediated immunity. In addition, notably higher expression levels of neural proteins (85%) and receptors (80%) were detected in the opioid group compared to the control group. This study suggests modulation of CNS homeostasis, possibly attributable to opioids, thus highlighting potential mechanisms for the pharmacodynamics of opioids. We also provide new insights into the immunomodulatory functions of opioids in vivo.
Collapse
|
20
|
Achour B, Al-Majdoub ZM, Rostami-Hodjegan A, Barber J. Mass Spectrometry of Human Transporters. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:223-247. [PMID: 32084322 DOI: 10.1146/annurev-anchem-091719-024553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Transporters are key to understanding how an individual will respond to a particular dose of a drug. Two patients with similar systemic concentrations may have quite different local concentrations of a drug at the required site. The transporter profile of any individual depends upon a variety of genetic and environmental factors, including genotype, age, and diet status. Robust models (virtual patients) are therefore required and these models are data hungry. Necessary data include quantitative transporter profiles at the relevant organ. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently the most powerful method available for obtaining this information. Challenges include sourcing the tissue, isolating the hydrophobic membrane-embedded transporter proteins, preparing the samples for MS (including proteolytic digestion), choosing appropriate quantification methodology, and optimizing the LC-MS/MS conditions. Great progress has been made with all of these, especially within the last few years, and is discussed here.
Collapse
Affiliation(s)
- Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
- Certara, Princeton, New Jersey 08540, USA
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
21
|
Bousalis D, Lacko CS, Hlavac N, Alkassis F, Wachs RA, Mobini S, Schmidt CE, Kasahara H. Extracellular Matrix Disparities in an Nkx2-5 Mutant Mouse Model of Congenital Heart Disease. Front Cardiovasc Med 2020; 7:93. [PMID: 32548129 PMCID: PMC7272573 DOI: 10.3389/fcvm.2020.00093] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/29/2020] [Indexed: 12/19/2022] Open
Abstract
Congenital heart disease (CHD) affects almost one percent of all live births. Despite diagnostic and surgical reparative advances, the causes and mechanisms of CHD are still primarily unknown. The extracellular matrix plays a large role in cell communication, function, and differentiation, and therefore likely plays a role in disease development and pathophysiology. Cell adhesion and gap junction proteins, such as integrins and connexins, are also essential to cellular communication and behavior, and could interact directly (integrins) or indirectly (connexins) with the extracellular matrix. In this work, we explore disparities in the expression and spatial patterning of extracellular matrix, adhesion, and gap junction proteins between wild type and Nkx2-5 +/R52G mutant mice. Decellularization and proteomic analysis, Western blotting, histology, immunostaining, and mechanical assessment of embryonic and neonatal wild type and Nkx2-5 mutant mouse hearts were performed. An increased abundance of collagen IV, fibronectin, and integrin β-1 was found in Nkx2-5 mutant neonatal mouse hearts, as well as increased expression of connexin 43 in embryonic mutant hearts. Furthermore, a ventricular noncompaction phenotype was observed in both embryonic and neonatal mutant hearts, as well as spatial disorganization of ECM proteins collagen IV and laminin in mutant hearts. Characterizing such properties in a mutant mouse model provides valuable information that can be applied to better understanding the mechanisms of congenital heart disease.
Collapse
Affiliation(s)
- Deanna Bousalis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Christopher S Lacko
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Nora Hlavac
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Fariz Alkassis
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| | - Rebecca A Wachs
- Department of Biological Systems Engineering, University of Nebraska-Lincoln, Lincoln, NE, United States
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM+CSIC), Madrid, Spain.,Centro de Biología Molecular Severo Ochoa (CBMSO, UAM-CSIC), Universidad Autónoma de Madrid, Madrid, Spain
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
22
|
Couto N, Al-Majdoub ZM, Gibson S, Davies PJ, Achour B, Harwood MD, Carlson G, Barber J, Rostami-Hodjegan A, Warhurst G. Quantitative Proteomics of Clinically Relevant Drug-Metabolizing Enzymes and Drug Transporters and Their Intercorrelations in the Human Small Intestine. Drug Metab Dispos 2020; 48:245-254. [PMID: 31959703 PMCID: PMC7076527 DOI: 10.1124/dmd.119.089656] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/23/2019] [Indexed: 01/02/2023] Open
Abstract
The levels of drug-metabolizing enzymes (DMEs) and transporter proteins in the human intestine are pertinent to determine oral drug bioavailability. Despite the paucity of reports on such measurements, it is well recognized that these values are essential for translating in vitro data on drug metabolism and transport to predict drug disposition in gut wall. In the current study, clinically relevant DMEs [cytochrome P450 (P450) and uridine 5'-diphospho-glucuronosyltransferase (UGT)] and drug transporters were quantified in total mucosal protein preparations from the human jejunum (n = 4) and ileum (n = 12) using quantification concatemer-based targeted proteomics. In contrast to previous reports, UGT2B15 and organic anion-transporting polypeptide 1 (OATP1A2) were quantifiable in all our samples. Overall, no significant disparities in protein expression were observed between jejunum and ileum. Relative mRNA expression for drug transporters did not correlate with the abundance of their cognate protein, except for P-glycoprotein 1 (P-gp) and organic solute transporter subunit alpha (OST-α), highlighting the limitations of RNA as a surrogate for protein expression in dynamic tissues with high turnover. Intercorrelations were found within P450 [2C9-2C19 (P = 0.002, R 2 = 0.63), 2C9-2J2 (P = 0.004, R 2 = 0.40), 2D6-2J2 (P = 0.002, R 2 = 0.50)] and UGT [1A1-2B7 (P = 0.02, R 2 = 0.87)] family of enzymes. There were also correlations between P-gp and several other proteins [OST-α (P < 0.0001, R 2 = 0.77), UGT1A6 (P = 0.009, R 2 = 0.38), and CYP3A4 (P = 0.007, R 2 = 0.30)]. Incorporating such correlations into building virtual populations is crucial for obtaining plausible characteristics of simulated individuals. SIGNIFICANCE STATEMENT: A number of drug transporters were quantified for the first time in this study. Several intercorrelations of protein abundance were reported. mRNA expression levels proved to be a poor reflection of differences between individuals regarding the level of protein expression in gut. The reported abundance of drug-metabolizing enzymes and transporters and their intercorrelations will contribute to better predictions of oral drug bioavailability and drug-drug interactions by linking in vitro observations to potential outcomes through physiologically based pharmacokinetic models.
Collapse
Affiliation(s)
- Narciso Couto
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Stephanie Gibson
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Pamela J Davies
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Matthew D Harwood
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Gordon Carlson
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| | - Geoffrey Warhurst
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.C., Z.M.A.-M., B.A., J.B., A.R.-H.); Gut Barrier Group, Inflammation and Repair, University of Manchester, Salford Royal NHS Trust, Salford, United Kingdom (S.G., P.J.D., G.C., G.W.); and Certara UK Limited (Simcyp Division), Sheffield, United Kingdom (M.D.H., A.R.-H.)
| |
Collapse
|