1
|
Huang D, Wu Z, Wang J, Wang J, Zhao Y. Biomimetic Liver Lobules from Multi-Compartmental Microfluidics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406573. [PMID: 39297364 PMCID: PMC11558095 DOI: 10.1002/advs.202406573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/21/2024] [Indexed: 11/14/2024]
Abstract
Engineered liver lobule is highly practical in hepatic disease treatment, while constructing a 3D biomimetic lobule with a heterogeneous architecture on a large scale is challenging. Here, inspired by the natural architectural construction of hepatic lobules, biomimetic hepatic lobules are proposed with coaxially through-pores for nutrient exchange via microfluidic technology. This multi-channel microfluidic chip is made by parallelly installing capillaries. Sodium alginate (Alg) is pumped through its central channel, while Ca2+-loaded gelatin methacrylate (GelMA) solutions encapsulating hepatocytes, mesenchymal stem cells, and endothelia cells are pumped through surrounding channels, respectively. The rapid gelation of Alg and Ca2+ brings about an in situ formation of Alg fiber, with heterogeneous multi-cell-laden GelMA microcarriers forming around it. The peeled-off microcarriers each featured with a coaxially through pore, simulating the cord-like structure of hepatic lobule and facilitating nutrients exchange. Meanwhile, the spatially anisotropic arrangement of cells highly simulates the hepatic architecture. It is demonstrated that by transplanting these biomimetic microparticles into liver in situ, the failed liver in rat shows increased regeneration and decreased necrosis. These results indicated that the microfluidic multi-compartmental microcarriers provide a new strategy to engineer 3D artificial livers for clinical translation.
Collapse
Affiliation(s)
- Danqing Huang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
| | - Zhuhao Wu
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
| | - Ji Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation SurgeryDepartment of General SurgeryNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjing210008China
- Shenzhen Research InstituteSoutheast UniversityShenzhen518071China
- Institute of Organoids on Chips Translational ResearchHenan Academy of SciencesZhengzhou450009China
| |
Collapse
|
2
|
Khojasteh SC, Argikar UA, Chatzopoulou M, Cheruzel L, Cho S, Dhaware D, Johnson KM, Kalgutkar AS, Liu J, Ma B, Maw H, Rowley JA, Seneviratne HK, Wang S. Biotransformation research advances - 2023 year in review. Drug Metab Rev 2024; 56:190-222. [PMID: 38989688 DOI: 10.1080/03602532.2024.2370330] [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: 03/27/2024] [Accepted: 06/14/2024] [Indexed: 07/12/2024]
Abstract
This annual review marks the eighth in the series starting with Baillie et al. (2016) Our objective is to explore and share articles which we deem influential and significant in the field of biotransformation. Its format is to highlight important aspects captured in synopsis followed by a commentary with relevant figure and references.
Collapse
Affiliation(s)
- S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Maria Chatzopoulou
- Early Clinical Development and Translational Science, UCB Biopharma UK, Slough, UK
| | - Lionel Cheruzel
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Sungjoon Cho
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | | | - Kevin M Johnson
- Drug Metabolism and Pharmacokinetics, Inotiv, MD Heights, MO, USA
| | - Amit S Kalgutkar
- Medicine Design, Pfizer Worldwide Research, Development and Medical, Cambridge, MA, USA
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| | - Hlaing Maw
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc, Ridgefield, CT, USA
| | - Jessica A Rowley
- Early Clinical Development and Translational Science, UCB Biopharma UK, Slough, UK
| | - Herana Kamal Seneviratne
- Department of Chemistry and Biochemistry, University of Maryland, Baltimore County, Baltimore, MD, USA
| | - Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
3
|
Wang S, Ballard TE, Christopher LJ, Foti RS, Gu C, Khojasteh SC, Liu J, Ma S, Ma B, Obach RS, Schadt S, Zhang Z, Zhang D. The Importance of Tracking "Missing" Metabolites: How and Why? J Med Chem 2023; 66:15586-15612. [PMID: 37769129 DOI: 10.1021/acs.jmedchem.3c01293] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Technologies currently employed to find and identify drug metabolites in complex biological matrices generally yield results that offer a comprehensive picture of the drug metabolite profile. However, drug metabolites can be missed or are captured only late in the drug development process. This could be due to a variety of factors, such as metabolism that results in partial loss of the molecule, covalent bonding to macromolecules, the drug being metabolized in specific human tissues, or poor ionization in a mass spectrometer. These scenarios often draw a great deal of attention from chemistry, safety assessment, and pharmacology. This review will summarize scenarios of missing metabolites, why they are missing, and associated uncovering strategies from deeper investigations. Uncovering previously missed metabolites can have ramifications in drug development with toxicological and pharmacological consequences, and knowledge of these can help in the design of new drugs.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - T Eric Ballard
- Takeda Development Center Americas, Inc., 35 Landsdowne St, Cambridge, Massachusetts 02139, United States
| | - Lisa J Christopher
- Department of Clinical Pharmacology, Pharmacometrics, Disposition & Bioanalysis, Bristol-Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Robert S Foti
- Preclinical Development, Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Chungang Gu
- Drug Metabolism and Pharmacokinetics, Biogen Inc., 225 Binney Street, Cambridge, Massachusetts 02142, United States
| | - S Cyrus Khojasteh
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joyce Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shuguang Ma
- Drug Metabolism and Pharmacokinetics, Pliant Therapeutics, 260 Littlefield Avenue, South San Francisco, California 94080, United States
| | - Bin Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - R Scott Obach
- Pharmacokinetics, Dynamics, and Metabolism, Pfizer, Inc., Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simone Schadt
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacher Strasse 124, 4070 Basel, Switzerland
| | - Zhoupeng Zhang
- DMPK Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Donglu Zhang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
4
|
Di L. Recent advances in measurement of metabolic clearance, metabolite profile and reaction phenotyping of low clearance compounds. Expert Opin Drug Discov 2023; 18:1209-1219. [PMID: 37526497 DOI: 10.1080/17460441.2023.2238606] [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: 04/05/2023] [Accepted: 07/17/2023] [Indexed: 08/02/2023]
Abstract
INTRODUCTION Low metabolic clearance is usually a highly desirable property of drug candidates in order to reduce dose and dosing frequency. However, measurement of low clearance can be challenging in drug discovery. A number of new tools have recently been developed to address the gaps in the measurement of intrinsic clearance, identification of metabolites, and reaction phenotyping of low clearance compounds. AREAS COVERED The new methodologies of low clearance measurements are discussed, including the hepatocyte relay, HepatoPac®, HμREL®, and spheroid systems. In addition, metabolite formation rate determination and in vivo allometric scaling approaches are covered as alternative methods for low clearance measurements. With these new methods, measurement of ~ 20-fold lower limit of intrinsic clearance can be achieved. The advantages and limitations of each approach are highlighted. EXPERT OPINION Although several novel methods have been developed in recent years to address the challenges of low clearance, these assays tend to be time and labor intensive and costly. Future innovations focusing on developing systems with high enzymatic activities, ultra-sensitive universal quantifiable detectors, and artificial intelligence will further enhance our ability to explore the low clearance space.
Collapse
Affiliation(s)
- Li Di
- Research Fellow, Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA
| |
Collapse
|
5
|
Moreau M, Mallick P, Smeltz M, Haider S, Nicolas CI, Pendse SN, Leonard JA, Linakis MW, McMullen PD, Clewell RA, Clewell HJ, Yoon M. Considerations for Improving Metabolism Predictions for In Vitro to In Vivo Extrapolation. FRONTIERS IN TOXICOLOGY 2022; 4:894569. [PMID: 35573278 PMCID: PMC9099212 DOI: 10.3389/ftox.2022.894569] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/13/2022] [Indexed: 12/14/2022] Open
Abstract
High-throughput (HT) in vitro to in vivo extrapolation (IVIVE) is an integral component in new approach method (NAM)-based risk assessment paradigms, for rapidly translating in vitro toxicity assay results into the context of in vivo exposure. When coupled with rapid exposure predictions, HT-IVIVE supports the use of HT in vitro assays for risk-based chemical prioritization. However, the reliability of prioritization based on HT bioactivity data and HT-IVIVE can be limited as the domain of applicability of current HT-IVIVE is generally restricted to intrinsic clearance measured primarily in pharmaceutical compounds. Further, current approaches only consider parent chemical toxicity. These limitations occur because current state-of-the-art HT prediction tools for clearance and metabolite kinetics do not provide reliable data to support HT-IVIVE. This paper discusses current challenges in implementation of IVIVE for prioritization and risk assessment and recommends a path forward for addressing the most pressing needs and expanding the utility of IVIVE.
Collapse
Affiliation(s)
- Marjory Moreau
- ScitoVation, LLC, Durham, NC, United States
- *Correspondence: Marjory Moreau,
| | | | | | | | | | | | - Jeremy A. Leonard
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, United States
| | | | | | | | | | | |
Collapse
|
6
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
7
|
Monckton CP, Brown GE, Khetani SR. Latest impact of engineered human liver platforms on drug development. APL Bioeng 2021; 5:031506. [PMID: 34286173 PMCID: PMC8286174 DOI: 10.1063/5.0051765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a leading cause of drug attrition, which is partly due to differences between preclinical animals and humans in metabolic pathways. Therefore, in vitro human liver models are utilized in biopharmaceutical practice to mitigate DILI risk and assess related mechanisms of drug transport and metabolism. However, liver cells lose phenotypic functions within 1–3 days in two-dimensional monocultures on collagen-coated polystyrene/glass, which precludes their use to model the chronic effects of drugs and disease stimuli. To mitigate such a limitation, bioengineers have adapted tools from the semiconductor industry and additive manufacturing to precisely control the microenvironment of liver cells. Such tools have led to the fabrication of advanced two-dimensional and three-dimensional human liver platforms for different throughput needs and assay endpoints (e.g., micropatterned cocultures, spheroids, organoids, bioprinted tissues, and microfluidic devices); such platforms have significantly enhanced liver functions closer to physiologic levels and improved functional lifetime to >4 weeks, which has translated to higher sensitivity for predicting drug outcomes and enabling modeling of diseased phenotypes for novel drug discovery. Here, we focus on commercialized engineered liver platforms and case studies from the biopharmaceutical industry showcasing their impact on drug development. We also discuss emerging multi-organ microfluidic devices containing a liver compartment that allow modeling of inter-tissue crosstalk following drug exposure. Finally, we end with key requirements for engineered liver platforms to become routine fixtures in the biopharmaceutical industry toward reducing animal usage and providing patients with safe and efficacious drugs with unprecedented speed and reduced cost.
Collapse
Affiliation(s)
- Chase P Monckton
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60607, USA
| |
Collapse
|
8
|
Asadi M, Khalili M, Lotfi H, Vaghefi Moghaddam S, Zarghami N, André H, Alizadeh E. Liver bioengineering: Recent trends/advances in decellularization and cell sheet technologies towards translation into the clinic. Life Sci 2021; 276:119373. [PMID: 33744324 DOI: 10.1016/j.lfs.2021.119373] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/03/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023]
Abstract
Development of novel technologies provides the best tissue constructs engineering and maximizes their therapeutic effects in regenerative therapy, especially for liver dysfunctions. Among the currently investigated approaches of tissue engineering, scaffold-based and scaffold-free tissues are widely suggested for liver regeneration. Analogs of liver acellular extracellular matrix (ECM) are utilized in native scaffolds to increase the self-repair and healing ability of organs. Native ECM analog could improve liver repairing through providing the supportive framework for cells and signaling molecules, exerting normal biomechanical, biochemical, and physiological signal complexes. Recently, innovative cell sheet technology is introduced as an alternative for conventional tissue engineering with the advantage of fewer scaffold restrictions and cell culture on a Thermo-Responsive Polymer Surface. These sheets release the layered cells through a temperature-controlled procedure without enzymatic digestion, while preserving the cell-ECM contacts and adhesive molecules on cell-cell junctions. In addition, several novelties have been introduced into the cell sheet and decellularization technologies to aid cell growth, instruct differentiation/angiogenesis, and promote cell migration. In this review, recent trends, advancements, and issues linked to translation into clinical practice are dissected and compared regarding the decellularization and cell sheet technologies for liver tissue engineering.
Collapse
Affiliation(s)
- Maryam Asadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Khalili
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajie Lotfi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Helder André
- Department of Clinical Neuroscience, St. Erik Eye Hospital, Karolinska Institute, 11282 Stockholm, Sweden
| | - Effat Alizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Huang D, Zhang X, Fu X, Zu Y, Sun W, Zhao Y. Liver spheroids on chips as emerging platforms for drug screening. ENGINEERED REGENERATION 2021. [DOI: 10.1016/j.engreg.2021.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
10
|
Berg EL, Denker SP, O'Mahony A. Development and Validation of Disease Assays for Phenotypic Screening. PHENOTYPIC DRUG DISCOVERY 2020. [DOI: 10.1039/9781839160721-00020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Screening in phenotypic assays is an important strategy for the discovery of innovative drugs and novel targets. Here we present key strategies for developing successful phenotypic screens and prosecuting phenotypic drug discovery (PDD) programs. Successful screens incorporate physiological relevance through the use of human cell types and assay designs that have (1) strong mechanistic connection to clinical outcomes and (2) strong biological justification for both efficacy and safety. In addition to guidance for designing successful screens, we also propose incorporation of specific counterscreens at an early point in the program. The suggested counterscreens are based on analysis of 1000s of drugs and drug candidates profiled through a large set of human-based phenotypic assays. These assays include cytotoxicity in human primary vascular endothelial cells, proliferation of endothelial cells, and proliferation of lymphocytes, all under specific activation conditions. These counterscreens form a generic screening funnel to triage a large fraction of early-stage hits, binning compounds into those with undesirable mechanisms (associated with acute toxicity), mechanisms with utility for oncology indications, and mechanisms useful for autoimmune indications. The application of this screening funnel offers a standardized and more predictable path for prosecuting PDD programs, reducing the risk of failure, and improving program timelines.
Collapse
Affiliation(s)
- Ellen L. Berg
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| | - Sheryl P. Denker
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| | - Alison O'Mahony
- Eurofins Discovery 111 Anza Blvd, Suite 414 Burlingame CA 94010 USA
| |
Collapse
|
11
|
Kamel A, Bowlin S, Hosea N, Arkilo D, Laurenza A. In Vitro Metabolism of Slowly Cleared G Protein-Coupled Receptor 139 Agonist TAK-041 Using Rat, Dog, Monkey, and Human Hepatocyte Models (HepatoPac): Correlation with In Vivo Metabolism. Drug Metab Dispos 2020; 49:121-132. [PMID: 33273044 DOI: 10.1124/dmd.120.000246] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/24/2020] [Indexed: 12/26/2022] Open
Abstract
Hepatic metabolism of low-clearance compound TAK-041 was studied in two different in vitro model systems using rat, dog, monkey, and human suspended cryopreserved hepatocytes and HepatoPac micropatterned coculture model primary hepatocytes. The aim of this work was to investigate the most appropriate system to assess the biotransformation of TAK-041, determine any notable species difference in the rate and in the extent of its metabolic pathways, and establish correlation with in vivo metabolism. TAK-041 exhibited very low turnover in suspended cryopreserved hepatocyte suspensions for all species, with no metabolites observed in human hepatocytes. However, incubations conducted for up to 14 days in the HepatoPac model resulted in more robust metabolic turnover. The major biotransformation pathways of TAK-041 proceed via hydroxylation on the benzene ring fused to the oxotriazine moiety and subsequent sulfate, glucuronide, and glutathione conjugation reactions. The glutathione conjugate of TAK-041 undergoes further downstream metabolism to produce the cysteine S-conjugate, which then undergoes N-acetylation to mercapturic acid and/or conversion to β-lyase-derived thiol metabolites. The minor biotransformation pathways include novel ring closure and hydrolysis, hydroxylation, oxidative N-dealkylation, and subsequent reduction. The HepatoPac model shows a notable species difference in the rate and in the extent of metabolic pathways of TAK-041, with dogs having the fastest metabolic clearance and humans the slowest. Furthermore, the model shows its suitability for establishing correlation with in vivo metabolism of low-turnover and extensively metabolized compounds such as TAK-041, displaying an extensive and unusual downstream sequential β-lyase-derived thiol metabolism in preclinical species and human. SIGNIFICANCE STATEMENT: This study investigated the most appropriate in vitro system to assess the biotransformation of the low-turnover and extensively metabolized compound TAK-041, determine any notable species difference in the rate and in the extent of its metabolic pathways, and establish correlation with in vivo metabolism. The HepatoPac model was identified and showed its suitability for species comparison and establishing correlation, with in vivo metabolism displaying an extensive and unusual downstream sequential β-lyase-derived thiol metabolism in preclinical species and human.
Collapse
Affiliation(s)
- Amin Kamel
- Global Drug Metabolism and Pharmacokinetics, Takeda California Inc., San Diego, California (A.K., S.B., N.H.) and Clinical and Neuroscience Therapeutic Area, Takeda Boston Inc., Boston, Massachusetts (D.A., A.L.)
| | - Steve Bowlin
- Global Drug Metabolism and Pharmacokinetics, Takeda California Inc., San Diego, California (A.K., S.B., N.H.) and Clinical and Neuroscience Therapeutic Area, Takeda Boston Inc., Boston, Massachusetts (D.A., A.L.)
| | - Natalie Hosea
- Global Drug Metabolism and Pharmacokinetics, Takeda California Inc., San Diego, California (A.K., S.B., N.H.) and Clinical and Neuroscience Therapeutic Area, Takeda Boston Inc., Boston, Massachusetts (D.A., A.L.)
| | - Dimitrios Arkilo
- Global Drug Metabolism and Pharmacokinetics, Takeda California Inc., San Diego, California (A.K., S.B., N.H.) and Clinical and Neuroscience Therapeutic Area, Takeda Boston Inc., Boston, Massachusetts (D.A., A.L.)
| | - Antonio Laurenza
- Global Drug Metabolism and Pharmacokinetics, Takeda California Inc., San Diego, California (A.K., S.B., N.H.) and Clinical and Neuroscience Therapeutic Area, Takeda Boston Inc., Boston, Massachusetts (D.A., A.L.)
| |
Collapse
|
12
|
Chen AX, Chhabra A, Song HHG, Fleming HE, Chen CS, Bhatia SN. Controlled Apoptosis of Stromal Cells to Engineer Human Microlivers. ADVANCED FUNCTIONAL MATERIALS 2020; 30:1910442. [PMID: 33776613 PMCID: PMC7996305 DOI: 10.1002/adfm.201910442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/29/2020] [Indexed: 05/02/2023]
Abstract
Engineered tissue models comprise a variety of multiplexed ensembles in which combinations of epithelial, stromal, and immune cells give rise to physiologic function. Engineering spatiotemporal control of cell-cell and cell-matrix interactions within these 3D multicellular tissues would represent a significant advance for tissue engineering. In this work, a new method, entitled CAMEO (Controlled Apoptosis in Multicellular tissues for Engineered Organogenesis) enables the non-invasive triggering of controlled apoptosis to eliminate genetically-engineered cells from a pre-established culture. Using this approach, the contribution of stromal cells to the phenotypic stability of primary human hepatocytes is examined. 3D hepatic microtissues, in which fibroblasts can enhance phenotypic stability and accelerate aggregation into spheroids, were found to rely only transiently on fibroblast interaction to support multiple axes of liver function, such as protein secretion and drug detoxification. Due to its modularity, CAMEO has the promise to be readily extendable to other applications that are tied to the complexity of 3D tissue biology, from understanding in vitro organoid models to building artificial tissue grafts.
Collapse
Affiliation(s)
- Amanda X Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arnav Chhabra
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - H-H Greco Song
- Harvard-MIT Program in Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Heather E Fleming
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Christopher S Chen
- Biological Design Center, Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Sangeeta N Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
13
|
Ballard TE, Kratochwil N, Cox LM, Moen MA, Klammers F, Ekiciler A, Goetschi A, Walter I. Simplifying the Execution of HepatoPac MetID Experiments: Metabolite Profile and Intrinsic Clearance Comparisons. Drug Metab Dispos 2020; 48:804-810. [PMID: 32623369 DOI: 10.1124/dmd.120.000013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/12/2020] [Indexed: 02/02/2023] Open
Abstract
The HepatoPac micropatterned coculture (MPCC) hepatocyte system has been shown to be an effective tool to investigate the qualitative human and preclinical species' metabolite profiles of new drug candidates. However, additional improvements to the overall study conditions and execution, layout, and human-donor count could be made. To that end, we have evaluated several ways to increase the amount of data one can generate per MPCC plate and how to more efficiently execute a MPCC study for the purpose of metabolite generation. Herein, we compare a set of compounds using single- and 10-donor pooled human MPCC hepatocytes. Intrinsic clearance and mean metabolic activities assessed by diverse enzyme markers were comparable between the single- and 10-donor pool. We have confirmed that the generated metabolite profiles were indistinguishable between the single- and 10-donor pool and also that rat MPCC can be performed at 400 µl media volume, which greatly simplifies study execution. Additional tips for successful study execution are also described. SIGNIFICANCE STATEMENT: When using the HepatoPac micropatterned coculture (MPCC) system, sometimes simple experimental condition variables or problematic plate designs can hamper productive study execution. We evaluated conditions to increase the amount of data one can generate per MPCC plate and, perhaps more importantly, execute that study more efficiently with less likelihood of error. We describe some of our key learnings, provide an examination of enzyme activity levels and clearance values, and provide some recommendations to simplify the execution of a HepatoPac experiment.
Collapse
Affiliation(s)
- T Eric Ballard
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - N Kratochwil
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Loretta M Cox
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - Mark A Moen
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - F Klammers
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Ekiciler
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - A Goetschi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| | - I Walter
- Pharmacokinetics, Dynamics and Metabolism, Pfizer, Inc., Groton, Connecticut (T.E.B., L.M.C., M.A.M.) and Drug Disposition and Safety, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, Basel, Switzerland (N.K., F.K., A.E., A.G., I.W.)
| |
Collapse
|
14
|
Fowler S, Chen WLK, Duignan DB, Gupta A, Hariparsad N, Kenny JR, Lai WG, Liras J, Phillips JA, Gan J. Microphysiological systems for ADME-related applications: current status and recommendations for system development and characterization. LAB ON A CHIP 2020; 20:446-467. [PMID: 31932816 DOI: 10.1039/c9lc00857h] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Over the last decade, progress has been made on the development of microphysiological systems (MPS) for absorption, distribution, metabolism, and excretion (ADME) applications. Central to this progress has been proof of concept data generated by academic and industrial institutions followed by broader characterization studies, which provide evidence for scalability and applicability to drug discovery and development. In this review, we describe some of the advances made for specific tissue MPS and outline the desired functionality for such systems, which are likely to make them applicable for practical use in the pharmaceutical industry. Single organ MPS platforms will be valuable for modelling tissue-specific functions. However, dynamic organ crosstalk, especially in the context of disease or toxicity, can only be obtained with the use of inter-linked MPS models which will enable scientists to address questions at the intersection of pharmacokinetics (PK) and efficacy, or PK and toxicity. In the future, successful application of MPS platforms that closely mimic human physiology may ultimately reduce the need for animal models to predict ADME outcomes and decrease the overall risk and cost associated with drug development.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharma Research and Early Development, F.Hoffmann-La Roche Ltd, Grenzacherstrasse 124, CH4070, Basel, Switzerland
| | | | - David B Duignan
- Department of Drug Metabolism, Pharmacokinetics & Bioanalysis, AbbVie Bioresearch Center, Worcester, Massachusetts 01605, USA
| | - Anshul Gupta
- Amgen Research, 360 Binney St, Cambridge, MA 02141, USA
| | - Niresh Hariparsad
- Department of Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, 50 Northern Ave, Boston, MA, USA
| | - Jane R Kenny
- DMPK, Genentech, 1 DNA Way, South San Francisco 94080, USA
| | | | - Jennifer Liras
- Medicine Design, Pfizer Inc, 1 Portland Ave, Cambridge, MA 02139, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb R&D, PO Box 4000, Princeton, NJ 08543-4000, USA.
| |
Collapse
|
15
|
Heydari Z, Najimi M, Mirzaei H, Shpichka A, Ruoss M, Farzaneh Z, Montazeri L, Piryaei A, Timashev P, Gramignoli R, Nussler A, Baharvand H, Vosough M. Tissue Engineering in Liver Regenerative Medicine: Insights into Novel Translational Technologies. Cells 2020; 9:E304. [PMID: 32012725 PMCID: PMC7072533 DOI: 10.3390/cells9020304] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 01/17/2020] [Accepted: 01/21/2020] [Indexed: 12/15/2022] Open
Abstract
Organ and tissue shortage are known as a crucially important public health problem as unfortunately a small percentage of patients receive transplants. In the context of emerging regenerative medicine, researchers are trying to regenerate and replace different organs and tissues such as the liver, heart, skin, and kidney. Liver tissue engineering (TE) enables us to reproduce and restore liver functions, fully or partially, which could be used in the treatment of acute or chronic liver disorders and/or generate an appropriate functional organ which can be transplanted or employed as an extracorporeal device. In this regard, a variety of techniques (e.g., fabrication technologies, cell-based technologies, microfluidic systems and, extracorporeal liver devices) could be applied in tissue engineering in liver regenerative medicine. Common TE techniques are based on allocating stem cell-derived hepatocyte-like cells or primary hepatocytes within a three-dimensional structure which leads to the improvement of their survival rate and functional phenotype. Taken together, new findings indicated that developing liver tissue engineering-based techniques could pave the way for better treatment of liver-related disorders. Herein, we summarized novel technologies used in liver regenerative medicine and their future applications in clinical settings.
Collapse
Affiliation(s)
- Zahra Heydari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran 1665659911, Iran
| | - Mustapha Najimi
- Laboratory of Pediatric Hepatology and Cell Therapy, Institute of Experimental & Clinical Research, Université Catholique de Louvain, B-1200 Brussels, Belgium;
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan 121135879, Iran;
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov University, 119146 Moscow, Russia; (A.S.); (P.T.)
| | - Marc Ruoss
- Siegfried Weller Institute for Trauma Research, University of Tübingen, 72076 Tübingen, Germany; (M.R.); (A.N.)
| | - Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
| | - Leila Montazeri
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran;
| | - Abbas Piryaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717443, Iran
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov University, 119146 Moscow, Russia; (A.S.); (P.T.)
- Department of Polymers and Composites, N.N.Semenov Institute of Chemical Physics, 117977 Moscow, Russia
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;
| | - Andreas Nussler
- Siegfried Weller Institute for Trauma Research, University of Tübingen, 72076 Tübingen, Germany; (M.R.); (A.N.)
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Developmental Biology, University of Science and Culture, ACECR, Tehran 1665659911, Iran
| | - Massoud Vosough
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran; (Z.H.); (Z.F.)
- Department of Regenerative Medicine, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665659911, Iran
| |
Collapse
|
16
|
Ware BR, Brown GE, Soldatow VY, LeCluyse EL, Khetani SR. Long-Term Engineered Cultures of Primary Mouse Hepatocytes for Strain and Species Comparison Studies During Drug Development. Gene Expr 2019; 19:199-214. [PMID: 31340881 PMCID: PMC6827040 DOI: 10.3727/105221619x15638857793317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Testing drugs in isogenic rodent strains to satisfy regulatory requirements is insufficient for derisking organ toxicity in genetically diverse human populations; in contrast, advances in mouse genetics can help mitigate these limitations. Compared to the expensive and slower in vivo testing, in vitro cultures enable the testing of large compound libraries toward prioritizing lead compounds and selecting an animal model with human-like response to a compound. In the case of the liver, a leading cause of drug attrition, isolated primary mouse hepatocytes (PMHs) rapidly decline in function within current culture platforms, which restricts their use for assessing the effects of longer-term compound exposure. Here we addressed this challenge by fabricating mouse micropatterned cocultures (mMPCC) containing PMHs and 3T3-J2 murine embryonic fibroblasts that displayed 4 weeks of functions; mMPCCs created from either C57Bl/6J or CD-1 PMHs outperformed collagen/Matrigel™ sandwich-cultured hepatocyte monocultures by ∼143-fold, 413-fold, and 10-fold for albumin secretion, urea synthesis, and cytochrome P450 activities, respectively. Such functional longevity of mMPCCs enabled in vivo relevant comparisons across strains for CYP induction and hepatotoxicity following exposure to 14 compounds with subsequent comparison to responses in primary human hepatocytes (PHHs). In conclusion, mMPCCs display high levels of major liver functions for several weeks and can be used to assess strain- and species-specific compound effects when used in conjunction with responses in PHHs. Ultimately, mMPCCs can be used to leverage the power of mouse genetics for characterizing subpopulations sensitive to compounds, characterizing the degree of interindividual variability, and elucidating genetic determinants of severe hepatotoxicity in humans.
Collapse
Affiliation(s)
- Brenton R. Ware
- *School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Grace E. Brown
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Edward L. LeCluyse
- ‡The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA
| | - Salman R. Khetani
- *School of Biomedical Engineering, Colorado State University, Fort Collins, CO, USA
- †Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
- §Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
17
|
Aleo MD, Ukairo O, Moore A, Irrechukwu O, Potter DM, Schneider RP. Liver safety evaluation of endothelin receptor antagonists using HepatoPac
®
: A single model impact assessment on hepatocellular health, function and bile acid disposition. J Appl Toxicol 2019; 39:1192-1207. [DOI: 10.1002/jat.3805] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Michael D. Aleo
- Drug Safety Research and Development, Worldwide Research & DevelopmentPfizer Inc. Groton Connecticut
| | | | - Amanda Moore
- BioIVT, formerly Hepregen Corporation Medford Massachusetts
| | | | - David M. Potter
- Drug Safety Research and Development, Worldwide Research & DevelopmentPfizer Inc. Groton Connecticut
| | - Richard P. Schneider
- Pharmacokinetics, Dynamics and Metabolism, Worldwide Research & DevelopmentPfizer Inc. Groton Connecticut
| |
Collapse
|
18
|
Brown GE, Khetani SR. Microfabrication of liver and heart tissues for drug development. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2017.0225. [PMID: 29786560 DOI: 10.1098/rstb.2017.0225] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/24/2017] [Indexed: 12/12/2022] Open
Abstract
Drug-induced liver- and cardiotoxicity remain among the leading causes of preclinical and clinical drug attrition, marketplace drug withdrawals and black-box warnings on marketed drugs. Unfortunately, animal testing has proven to be insufficient for accurately predicting drug-induced liver- and cardiotoxicity across many drug classes, likely due to significant differences in tissue functions across species. Thus, the field of in vitro human tissue engineering has gained increasing importance over the last 10 years. Technologies such as protein micropatterning, microfluidics, three-dimensional scaffolds and bioprinting have revolutionized in vitro platforms as well as increased the long-term phenotypic stability of both primary cells and stem cell-derived differentiated cells. Here, we discuss advances in engineering approaches for constructing in vitro human liver and heart models with utility for drug development. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues. Overall, bioengineered liver and heart models have significantly advanced our understanding of organ function and injury, which will prove useful for mitigating the risk of drug-induced organ toxicity to human patients, reducing animal usage for preclinical drug testing, aiding in the discovery of novel therapeutics against human diseases, and ultimately for applications in regenerative medicine.This article is part of the theme issue 'Designer human tissue: coming to a lab near you'.
Collapse
Affiliation(s)
- Grace E Brown
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
19
|
Mancio-Silva L, Fleming HE, Miller AB, Milstein S, Liebow A, Haslett P, Sepp-Lorenzino L, Bhatia SN. Improving Drug Discovery by Nucleic Acid Delivery in Engineered Human Microlivers. Cell Metab 2019; 29:727-735.e3. [PMID: 30840913 PMCID: PMC6408324 DOI: 10.1016/j.cmet.2019.02.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/21/2018] [Accepted: 02/11/2019] [Indexed: 12/20/2022]
Abstract
The liver plays a central role in metabolism; however, xenobiotic metabolism variations between human hepatocytes and those in model organisms create challenges in establishing functional test beds to detect the potential drug toxicity and efficacy of candidate small molecules. In the emerging areas of RNA interference, viral gene therapy, and genome editing, more robust, long-lasting, and predictive human liver models may accelerate progress. Here, we apply a new modality to a previously established, functionally stable, multi-well bioengineered microliver-fabricated from primary human hepatocytes and supportive stromal cells-in order to advance both small molecule and nucleic acid therapeutic pipelines. Specifically, we achieve robust and durable gene silencing in vitro to tune the human metabolism of small molecules, and demonstrate its capacity to query the potential efficacy and/or toxicity of candidate therapeutics. Additionally, we apply this engineered platform to test siRNAs designed to target hepatocytes and impact human liver genetic and infectious diseases.
Collapse
Affiliation(s)
- Liliana Mancio-Silva
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Heather E Fleming
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Alex B Miller
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Stuart Milstein
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Abigail Liebow
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | - Patrick Haslett
- Alnylam Pharmaceuticals, 300 3rd Street, Cambridge, MA 02142, USA
| | | | - Sangeeta N Bhatia
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA; Koch Institute for Integrative Cancer Research, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Docci L, Parrott N, Krähenbühl S, Fowler S. Application of New Cellular and Microphysiological Systems to Drug Metabolism Optimization and Their Positioning Respective to In Silico Tools. SLAS DISCOVERY 2019; 24:523-536. [PMID: 30817893 DOI: 10.1177/2472555219831407] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
New cellular model systems for drug metabolism applications, such as advanced 2D liver co-cultures, spheroids, and microphysiological systems (MPSs), offer exciting opportunities to reproduce human biology more closely in vitro with the aim of improving predictions of pharmacokinetics, drug-drug interactions, and efficacy. These advanced cellular systems have quickly become established for human intrinsic clearance determination and have been validated for several other absorption, distribution, metabolism, and excretion (ADME) applications. Adoption will be driven through the demonstration of clear added value, for instance, by more accurate and precise clearance predictions and by more reliable extrapolation of drug interaction potential leading to faster progression to pivotal proof-of-concept studies. New experimental systems are attractive when they can (1) increase experimental capacity, removing optimization bottlenecks; (2) improve measurement quality of ADME properties that impact pharmacokinetics; and (3) enable measurements to be made that were not previously possible, reducing risk in ADME prediction and candidate selection. As new systems become established, they will find their place in the repository of tools used at different stages of the research and development process, depending on the balance of value, throughput, and cost. In this article, we give a perspective on the integration of these new methodologies into ADME optimization during drug discovery, and the likely applications and impacts on drug development.
Collapse
Affiliation(s)
- Luca Docci
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland.,2 Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Neil Parrott
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| | | | - Stephen Fowler
- 1 Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
21
|
Xie J, Saburulla NF, Chen S, Wong SY, Yap ZP, Zhang LH, Lau AJ. Evaluation of Carbazeran 4-Oxidation and O6-Benzylguanine 8-Oxidation as Catalytic Markers of Human Aldehyde Oxidase: Impact of Cytosolic Contamination of Liver Microsomes. Drug Metab Dispos 2018; 47:26-37. [DOI: 10.1124/dmd.118.082099] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 10/15/2018] [Indexed: 12/17/2022] Open
|
22
|
Phillips MB, Balbuena-Venancio P, Enders JR, Norini RL, Shim YS, Burgunder E, Rao L, Billings D, Pedersen J, Macdonald JM, Andersen M, Clewell HJ, Yoon M. Xenobiotic Metabolism in Alginate-Encapsulated Primary Human Hepatocytes Over Long Timeframes. ACTA ACUST UNITED AC 2018. [DOI: 10.1089/aivt.2017.0029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
| | - Pergentino Balbuena-Venancio
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | | | | | - Yoo-Sik Shim
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Erin Burgunder
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Lavanya Rao
- ScitoVation, LLC, Research Triangle Park, North Carolina
| | - David Billings
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Jenny Pedersen
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Jeffrey M. Macdonald
- Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, North Carolina
| | - Melvin Andersen
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Harvey J. Clewell
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| | - Miyoung Yoon
- ScitoVation, LLC, Research Triangle Park, North Carolina
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, North Carolina
| |
Collapse
|
23
|
Underhill GH, Khetani SR. Advances in Engineered Human Liver Platforms for Drug Metabolism Studies. Drug Metab Dispos 2018; 46:1626-1637. [PMID: 30135245 DOI: 10.1124/dmd.118.083295] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022] Open
Abstract
Metabolism in the liver often determines the overall clearance rates of many pharmaceuticals. Furthermore, induction or inhibition of the liver drug metabolism enzymes by perpetrator drugs can influence the metabolism of victim drugs (drug-drug interactions). Therefore, determining liver-drug interactions is critical during preclinical drug development. Unfortunately, studies in animals are often of limited value because of significant differences in the metabolic pathways of the liver across different species. To mitigate such limitations, the pharmaceutical industry uses a continuum of human liver models, ranging from microsomes to transfected cell lines and cultures of primary human hepatocytes (PHHs). Of these models, PHHs provide a balance of high-throughput testing capabilities together with a physiologically relevant cell type that exhibits all the characteristic enzymes, cofactors, and transporters. However, PHH monocultures display a rapid decline in metabolic capacity. Consequently, bioengineers have developed several tools, such as cellular microarrays, micropatterned cocultures, self-assembled and bioprinted spheroids, and perfusion devices, to enhance and stabilize PHH functions for ≥2 weeks. Many of these platforms have been validated for drug studies, whereas some have been adapted to include liver nonparenchymal cells that can influence hepatic drug metabolism in health and disease. Here, we focus on the design features of such platforms and their representative drug metabolism validation datasets, while discussing emerging trends. Overall, the use of engineered human liver platforms in the pharmaceutical industry has been steadily rising over the last 10 years, and we anticipate that these platforms will become an integral part of drug development with continued commercialization and validation for routine screening use.
Collapse
Affiliation(s)
- Gregory H Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| | - Salman R Khetani
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois; and Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
24
|
Burton RD, Hieronymus T, Chamem T, Heim D, Anderson S, Zhu X, Hutzler JM. Assessment of the Biotransformation of Low-Turnover Drugs in the H µREL Human Hepatocyte Coculture Model. Drug Metab Dispos 2018; 46:1617-1625. [PMID: 30135244 DOI: 10.1124/dmd.118.082867] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/15/2018] [Indexed: 01/24/2023] Open
Abstract
Metabolic profiles of four drugs possessing diverse metabolic pathways (timolol, meloxicam, linezolid, and XK469) were compared following incubations in both suspended cryopreserved human hepatocytes and the HμREL hepatocyte coculture model. In general, minimal metabolism was observed following 4-hour incubations in both suspended hepatocytes and the HμREL model, whereas incubations conducted up to 7 days in the HμREL coculture model resulted in more robust metabolic turnover. In the case of timolol, in vivo human data suggest that 22% of the dose is transformed via multistep oxidative opening of the morpholine moiety. Only the first-step oxidation was detected in suspended hepatocytes, whereas the relevant downstream metabolites were produced in the HµREL model. For meloxicam, both the hydroxymethyl and subsequent carboxylic acid metabolites were abundant following incubation in the HμREL model, while only a trace amount of the hydroxymethyl metabolite was observed in suspension. Similar to timolol, linezolid generated substantially higher levels of morpholine ring-opened carboxylic acid metabolites in the HμREL model. Finally, while the major aldehyde oxidase-mediated mono-oxidative metabolite of XK469 was minimally produced in hepatocyte suspension, the HμREL model robustly produced this metabolite, consistent with a pathway reported to account for 54% of the total urinary excretion in human. In addition, low-level taurine and glycine conjugates were identified in the HµREL model. In summary, continuous metabolite production was observed for up to 7 days of incubation in the HµREL model, covering cytochrome P450, aldehyde oxidase, and numerous conjugative pathways, while predominant metabolites correlated with relevant metabolites reported in human in vivo studies.
Collapse
Affiliation(s)
- Richard D Burton
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Todd Hieronymus
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Taysir Chamem
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - David Heim
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Shelby Anderson
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - Xiaochun Zhu
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| | - J Matthew Hutzler
- Q Solutions, a Quintiles Quest Joint Venture, Bioanalytical and ADME Laboratories, Indianapolis, Indiana
| |
Collapse
|
25
|
Caetano-Pinto P, Stahl SH. Perspective on the Application of Microphysiological Systems to Drug Transporter Studies. Drug Metab Dispos 2018; 46:1647-1657. [DOI: 10.1124/dmd.118.082750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022] Open
|
26
|
Zhu Y, Liu W, Qi S, Wang H, Wang Y, Deng G, Zhang Y, Li S, Ma C, Wang Y, Cheng X, Wang C. Stereoselective glucuronidation metabolism, pharmacokinetics, anti-amnesic pharmacodynamics, and toxic properties of vasicine enantiomers in vitro and in vivo. Eur J Pharm Sci 2018; 123:459-474. [PMID: 30077712 DOI: 10.1016/j.ejps.2018.07.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 07/24/2018] [Accepted: 07/30/2018] [Indexed: 01/05/2023]
Abstract
Vasicine (VAS) is a potential natural cholinesterase inhibitor for treatment of Alzheimer's disease. Due to one chiral centre (C-3) presenting in molecule, VAS has two enantiomers, d-vasicine (d-VAS) and l-vasicine (l-VAS). The study was undertaken to investigate the stereoselective glucuronidation metabolism, pharmacokinetics, anti-amnesic effect and acute toxicity of VAS enantiomers. In results, the glucuronidation metabolic rate of l-VAS was faster than d-VAS in human liver microsomes and isoenzymes tests, and it was proved that the UDP-glucuronosyltransferase (UGT) 1A9 and UGT2B15 were the major metabolic enzymes for glucuronidation of l-VAS, while only UGT1A9 for d-VAS, which take responsibility of the significantly less metabolic affinity of d-VAS than l-VAS in HLM and rhUGT1A9. The plasma exposure of d-VAS in rats was 1.3-fold and 1.6-fold higher than that of l-VAS after intravenous and oral administration of d-VAS and l-VAS, respectively. And the plasma exposure of the major glucuronidation metabolite d-VASG was one of tenth of l-VASG or more less, no matter by intravenous or oral administration. Both d-VAS and l-VAS were exhibited promising acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) inhibitory activities, and the BChE inhibitory activity of d-VAS with IC50 of 0.03 ± 0.001 μM was significantly stronger than that of l-VAS with IC50 of 0.98 ± 0.19 μM. The molecular docking results indicated that d-VAS and l-VAS could bind to the catalytic active site (CAS position) either of human AChE and BChE, and the BChE combing ability of d-VAS (the score of GBI/WAS dG -7.398) was stronger than that of l-VAS (the score of GBI/WAS dG -7.135). Both d-VAS and l-VAS could improving the learning and memory on scopolamine-induced memory deficits in mice. The content of acetylcholine (ACh) after oral administration d-VAS increased more than that of l-VAS in mice cortex, through inhibiting cholinesterase (ChE) and increasing choline acetyltransferase (ChAT). In addition, the LD50 value of d-VAS (282.51 mg·kg-1) was slight lower than l-VAS (319.75 mg·kg-1). These results indicated that VAS enantiomers displayed significantly stereoselective metabolic, pharmacokinetics, anti-amnesic effect and toxic properties in vitro and in vivo. The d-VAS might be the dominant configuration for treating Alzheimer's disease.
Collapse
Affiliation(s)
- Yudan Zhu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Wei Liu
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Shenglan Qi
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Hanxue Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Yuwen Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Gang Deng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Yunpeng Zhang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Shuping Li
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Chao Ma
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Yongli Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Xuemei Cheng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China
| | - Changhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, The MOE Key Laboratory for Standardization of Chinese Medicines, The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China; Shanghai R&D Centre for Standardization of Chinese Medicines, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
27
|
Fraser K, Bruckner DM, Dordick JS. Advancing Predictive Hepatotoxicity at the Intersection of Experimental, in Silico, and Artificial Intelligence Technologies. Chem Res Toxicol 2018; 31:412-430. [PMID: 29722533 DOI: 10.1021/acs.chemrestox.8b00054] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Adverse drug reactions, particularly those that result in drug-induced liver injury (DILI), are a major cause of drug failure in clinical trials and drug withdrawals. Hepatotoxicity-mediated drug attrition occurs despite substantial investments of time and money in developing cellular assays, animal models, and computational models to predict its occurrence in humans. Underperformance in predicting hepatotoxicity associated with drugs and drug candidates has been attributed to existing gaps in our understanding of the mechanisms involved in driving hepatic injury after these compounds perfuse and are metabolized by the liver. Herein we assess in vitro, in vivo (animal), and in silico strategies used to develop predictive DILI models. We address the effectiveness of several two- and three-dimensional in vitro cellular methods that are frequently employed in hepatotoxicity screens and how they can be used to predict DILI in humans. We also explore how humanized animal models can recapitulate human drug metabolic profiles and associated liver injury. Finally, we highlight the maturation of computational methods for predicting hepatotoxicity, the untapped potential of artificial intelligence for improving in silico DILI screens, and how knowledge acquired from these predictions can shape the refinement of experimental methods.
Collapse
Affiliation(s)
- Keith Fraser
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Dylan M Bruckner
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| | - Jonathan S Dordick
- Department of Chemical and Biological Engineering and Department of Biological Sciences Center for Biotechnology and Interdisciplinary Studies , Rensselaer Polytechnic Institute , Troy , New York 12180 , United States
| |
Collapse
|
28
|
Underhill GH, Khetani SR. Bioengineered Liver Models for Drug Testing and Cell Differentiation Studies. Cell Mol Gastroenterol Hepatol 2018; 5:426-439.e1. [PMID: 29675458 PMCID: PMC5904032 DOI: 10.1016/j.jcmgh.2017.11.012] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/21/2017] [Indexed: 12/19/2022]
Abstract
In vitro models of the human liver are important for the following: (1) mitigating the risk of drug-induced liver injury to human beings, (2) modeling human liver diseases, (3) elucidating the role of single and combinatorial microenvironmental cues on liver cell function, and (4) enabling cell-based therapies in the clinic. Methods to isolate and culture primary human hepatocytes (PHHs), the gold standard for building human liver models, were developed several decades ago; however, PHHs show a precipitous decline in phenotypic functions in 2-dimensional extracellular matrix-coated conventional culture formats, which does not allow chronic treatment with drugs and other stimuli. The development of several engineering tools, such as cellular microarrays, protein micropatterning, microfluidics, biomaterial scaffolds, and bioprinting, now allow precise control over the cellular microenvironment for enhancing the function of both PHHs and induced pluripotent stem cell-derived human hepatocyte-like cells; long-term (4+ weeks) stabilization of hepatocellular function typically requires co-cultivation with liver-derived or non-liver-derived nonparenchymal cell types. In addition, the recent development of liver organoid culture systems can provide a strategy for the enhanced expansion of therapeutically relevant cell types. Here, we discuss advances in engineering approaches for constructing in vitro human liver models that have utility in drug screening and for determining microenvironmental determinants of liver cell differentiation/function. Design features and validation data of representative models are presented to highlight major trends followed by the discussion of pending issues that need to be addressed. Overall, bioengineered liver models have significantly advanced our understanding of liver function and injury, which will prove useful for drug development and ultimately cell-based therapies.
Collapse
Key Words
- 3D, 3-dimensional
- BAL, bioartificial liver
- Bioprinting
- CRP, C-reactive protein
- CYP450, cytochrome P450
- Cellular Microarrays
- DILI, drug-induced liver injury
- ECM, extracellular matrix
- HSC, hepatic stellate cell
- Hepatocytes
- IL, interleukin
- KC, Kupffer cell
- LSEC, liver sinusoidal endothelial cell
- MPCC, micropatterned co-culture
- Microfluidics
- Micropatterned Co-Cultures
- NPC, nonparenchymal cell
- PEG, polyethylene glycol
- PHH, primary human hepatocyte
- Spheroids
- iHep, induced pluripotent stem cell-derived human hepatocyte-like cell
- iPS, induced pluripotent stem
Collapse
Affiliation(s)
- Gregory H. Underhill
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Salman R. Khetani
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
29
|
Kratochwil NA, Triyatni M, Mueller MB, Klammers F, Leonard B, Turley D, Schmaler J, Ekiciler A, Molitor B, Walter I, Gonsard PA, Tournillac CA, Durrwell A, Marschmann M, Jones R, Ullah M, Boess F, Ottaviani G, Jin Y, Parrott NJ, Fowler S. Simultaneous Assessment of Clearance, Metabolism, Induction, and Drug-Drug Interaction Potential Using a Long-Term In Vitro Liver Model for a Novel Hepatitis B Virus Inhibitor. J Pharmacol Exp Ther 2018; 365:237-248. [PMID: 29453199 DOI: 10.1124/jpet.117.245712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/26/2018] [Indexed: 01/04/2023] Open
Abstract
Long-term in vitro liver models are now widely explored for human hepatic metabolic clearance prediction, enzyme phenotyping, cross-species metabolism, comparison of low clearance drugs, and induction studies. Here, we present studies using a long-term liver model, which show how metabolism and active transport, drug-drug interactions, and enzyme induction in healthy and diseased states, such as hepatitis B virus (HBV) infection, may be assessed in a single test system to enable effective data integration for physiologically based pharmacokinetic (PBPK) modeling. The approach is exemplified in the case of (3S)-4-[[(4R)-4-(2-Chloro-4-fluorophenyl)-5-methoxycarbonyl-2-thiazol-2-yl-1,4-dihydropyrimidin-6-yl]methyl]morpholine-3-carboxylic acid RO6889678, a novel inhibitor of HBV with a complex absorption, distribution, metabolism, and excretion (ADME) profile. RO6889678 showed an intracellular enrichment of 78-fold in hepatocytes, with an apparent intrinsic clearance of 5.2 µl/min per mg protein and uptake and biliary clearances of 2.6 and 1.6 µl/min per mg protein, respectively. When apparent intrinsic clearance was incorporated into a PBPK model, the simulated oral human profiles were in good agreement with observed data at low doses but were underestimated at high doses due to unexpected overproportional increases in exposure with dose. In addition, the induction potential of RO6889678 on cytochrome P450 (P450) enzymes and transporters at steady state was assessed and cotreatment with ritonavir revealed a complex drug-drug interaction with concurrent P450 inhibition and moderate UDP-glucuronosyltransferase induction. Furthermore, we report on the first evaluation of in vitro pharmacokinetics studies using HBV-infected HepatoPac cocultures. Thus, long-term liver models have great potential as translational research tools exploring pharmacokinetics of novel drugs in vitro in health and disease.
Collapse
Affiliation(s)
- Nicole A Kratochwil
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Miriam Triyatni
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Martina B Mueller
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Florian Klammers
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Brian Leonard
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Dan Turley
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Josephine Schmaler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Aynur Ekiciler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Birgit Molitor
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Isabelle Walter
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Pierre-Alexis Gonsard
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Charles A Tournillac
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Alexandre Durrwell
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Michaela Marschmann
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Russell Jones
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Mohammed Ullah
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Franziska Boess
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Giorgio Ottaviani
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Yuyan Jin
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Neil J Parrott
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| | - Stephen Fowler
- Pharmaceutical Sciences (N.A.K., M.B.M., F.K., A.E., B.M., I.W., P.-A.G., C.A.T., A.D., M.M., R.J., M.U., F.B., N.J.P., S.F.) and Inflammation, Immunology, and Infectious Diseases Therapeutic Areas (M.T., B.L., D.T., J.S.), Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland; and Pharmaceutical Sciences, Roche Innovation Center Shanghai, Roche R&D Center (China) Ltd., Pudong, Shanghai, China (G.O., Y.Y.)
| |
Collapse
|
30
|
Cassidy KC, Yi P. Qualitative and quantitative prediction of human in vivo metabolic pathways in a human hepatocyte-murine stromal cell co-culture model. Xenobiotica 2017; 48:1192-1205. [DOI: 10.1080/00498254.2017.1395927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
| | - Ping Yi
- Eli Lilly and Company, Lilly Corporate Centre , Indianapolis, IN , USA
| |
Collapse
|
31
|
Abstract
To curb the high cost of drug development, there is an urgent need to develop more predictive tissue models using human cells to determine drug efficacy and safety in advance of clinical testing. Recent insights gained through fundamental biological studies have validated the importance of dynamic cell environments and cellular communication to the expression of high fidelity organ function. Building on this knowledge, emerging organ-on-a-chip technology is poised to fill the gaps in drug screening by offering predictive human tissue models with methods of sophisticated tissue assembly. Organ-on-a-chip start-ups have begun to spawn from academic research to fill this commercial space and are attracting investment to transform the drug discovery industry. This review traces the history, examines the scientific foundation and envisages the prospect of these renowned organ-on-a-chip technologies. It serves as a guide for new members of this dynamic field to navigate the existing scientific and market space.
Collapse
Affiliation(s)
- Boyang Zhang
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, ON, Canada.
| | | |
Collapse
|
32
|
Sato-Nakai M, Kawashima K, Nakagawa T, Tachibana Y, Yoshida M, Takanashi K, Morcos PN, Binder M, Moore DJ, Yu L. Metabolites of alectinib in human: their identification and pharmacological activity. Heliyon 2017; 3:e00354. [PMID: 28725874 PMCID: PMC5506877 DOI: 10.1016/j.heliyon.2017.e00354] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 06/21/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023] Open
Abstract
Two metabolites (M4 and M1b) in plasma and four metabolites (M4, M6, M1a and M1b) in faeces were detected through the human ADME study following a single oral administration of [14C]alectinib, a small-molecule anaplastic lymphoma kinase inhibitor, to healthy subjects. In the present study, M1a and M1b, which chemical structures had not been identified prior to the human ADME study, were identified as isomers of a carboxylate metabolite oxidatively cleaved at the morpholine ring. In faeces, M4 and M1b were the main metabolites, which shows that the biotransformation to M4 and M1b represents two main metabolic pathways for alectinib. In plasma, M4 was a major metabolite and M1b was a minor metabolite. The contribution to in vivo pharmacological activity of these circulating metabolites was assessed from their in vitro pharmacological activity and plasma protein binding. M4 had a similar cancer cell growth inhibitory activity and plasma protein binding to that of alectinib, suggesting its contribution to the antitumor activity of alectinib, whereas the pharmacological activity of M1b was insignificant.
Collapse
Affiliation(s)
- Mika Sato-Nakai
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kosuke Kawashima
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Toshito Nakagawa
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Yukako Tachibana
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Miyuki Yoshida
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Kenji Takanashi
- Research division, Chugai Pharmaceuticals, Co., Ltd., 1-135 Komakado, Gotemba, Shizuoka 412-8513, Japan
| | - Peter N Morcos
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| | - Martin Binder
- Roche Innovation Center Basel, Knozern-Hauptsitz, Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - David J Moore
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| | - Li Yu
- Roche Innovation Center New York, 430 East 29th Street, New York, NY10016, United States
| |
Collapse
|
33
|
Takahashi RH, Shahidi-Latham SK, Wong S, Chang JH. Applying Stable Isotope Labeled Amino Acids in Micropatterned Hepatocyte Coculture to Directly Determine the Degradation Rate Constant for CYP3A4. Drug Metab Dispos 2017; 45:581-585. [DOI: 10.1124/dmd.116.074393] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 03/10/2017] [Indexed: 11/22/2022] Open
|
34
|
Fowler S, Morcos PN, Cleary Y, Martin-Facklam M, Parrott N, Gertz M, Yu L. Progress in Prediction and Interpretation of Clinically Relevant Metabolic Drug-Drug Interactions: a Minireview Illustrating Recent Developments and Current Opportunities. CURRENT PHARMACOLOGY REPORTS 2017; 3:36-49. [PMID: 28261547 PMCID: PMC5315728 DOI: 10.1007/s40495-017-0082-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW This review gives a perspective on the current "state of the art" in metabolic drug-drug interaction (DDI) prediction. We highlight areas of successful prediction and illustrate progress in areas where limits in scientific knowledge or technologies prevent us from having full confidence. RECENT FINDINGS Several examples of success are highlighted. Work done for bitopertin shows how in vitro and clinical data can be integrated to give a model-based understanding of pharmacokinetics and drug interactions. The use of interpolative predictions to derive explicit dosage recommendations for untested DDIs is discussed using the example of ibrutinib, and the use of DDI predictions in lieu of clinical studies in new drug application packages is exemplified with eliglustat and alectinib. Alectinib is also an interesting case where dose adjustment is unnecessary as the activity of a major metabolite compensates sufficiently for changes in parent drug exposure. Examples where "unusual" cytochrome P450 (CYP) and non-CYP enzymes are responsible for metabolic clearance have shown the importance of continuing to develop our repertoire of in vitro regents and techniques. The time-dependent inhibition assay using human hepatocytes suspended in full plasma allowed improved DDI predictions, illustrating the importance of continued in vitro assay development and refinement. SUMMARY During the past 10 years, a highly mechanistic understanding has been developed in the area of CYP-mediated metabolic DDIs enabling the prediction of clinical outcome based on preclinical studies. The combination of good quality in vitro data and physiologically based pharmacokinetic modeling may now be used to evaluate DDI risk prospectively and are increasingly accepted in lieu of dedicated clinical studies.
Collapse
Affiliation(s)
- Stephen Fowler
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Peter N. Morcos
- Pharmaceutical Reseach and Early Development, Roche Innovation Center New York, F. Hoffmann-La Roche Ltd., 430 East 29th Street, New York City, NY USA
| | - Yumi Cleary
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Meret Martin-Facklam
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Neil Parrott
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Michael Gertz
- Pharmaceutical Research and Early Development, Roche Innovation Centre Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - Li Yu
- Pharmaceutical Reseach and Early Development, Roche Innovation Center New York, F. Hoffmann-La Roche Ltd., 430 East 29th Street, New York City, NY USA
| |
Collapse
|
35
|
Sistare FD, Mattes WB, LeCluyse EL. The Promise of New Technologies to Reduce, Refine, or Replace Animal Use while Reducing Risks of Drug Induced Liver Injury in Pharmaceutical Development. ILAR J 2017; 57:186-211. [DOI: 10.1093/ilar/ilw025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
|
36
|
Pedersen JM, Shim YS, Hans V, Phillips MB, Macdonald JM, Walker G, Andersen ME, Clewell HJ, Yoon M. Fluid Dynamic Modeling to Support the Development of Flow-Based Hepatocyte Culture Systems for Metabolism Studies. Front Bioeng Biotechnol 2016; 4:72. [PMID: 27747210 PMCID: PMC5044513 DOI: 10.3389/fbioe.2016.00072] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/05/2016] [Indexed: 11/13/2022] Open
Abstract
Accurate prediction of metabolism is a significant outstanding challenge in toxicology. The best predictions are based on experimental data from in vitro systems using primary hepatocytes. The predictivity of the primary hepatocyte-based culture systems, however, is still limited due to well-known phenotypic instability and rapid decline of metabolic competence within a few hours. Dynamic flow bioreactors for three-dimensional cell cultures are thought to be better at recapitulating tissue microenvironments and show potential to improve in vivo extrapolations of chemical or drug toxicity based on in vitro test results. These more physiologically relevant culture systems hold potential for extending metabolic competence of primary hepatocyte cultures as well. In this investigation, we used computational fluid dynamics to determine the optimal design of a flow-based hepatocyte culture system for evaluating chemical metabolism in vitro. The main design goals were (1) minimization of shear stress experienced by the cells to maximize viability, (2) rapid establishment of a uniform distribution of test compound in the chamber, and (3) delivery of sufficient oxygen to cells to support aerobic respiration. Two commercially available flow devices – RealBio® and QuasiVivo® (QV) – and a custom developed fluidized bed bioreactor were simulated, and turbulence, flow characteristics, test compound distribution, oxygen distribution, and cellular oxygen consumption were analyzed. Experimental results from the bioreactors were used to validate the simulation results. Our results indicate that maintaining adequate oxygen supply is the most important factor to the long-term viability of liver bioreactor cultures. Cell density and system flow patterns were the major determinants of local oxygen concentrations. The experimental results closely corresponded to the in silico predictions. Of the three bioreactors examined in this study, we were able to optimize the experimental conditions for long-term hepatocyte cell culture using the QV bioreactor. This system facilitated the use of low system volumes coupled with higher flow rates. This design supports cellular respiration by increasing oxygen concentrations in the vicinity of the cells and facilitates long-term kinetic studies of low clearance test compounds. These two goals were achieved while simultaneously keeping the shear stress experienced by the cells within acceptable limits.
Collapse
Affiliation(s)
- Jenny M Pedersen
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences , Research Triangle Park, NC , USA
| | - Yoo-Sik Shim
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA; ScitoVation, LLC, Research Triangle Park, NC, USA; Joint Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, USA
| | - Vaibhav Hans
- Joint Department of Biomedical Engineering, University of North Carolina , Chapel Hill, NC , USA
| | | | - Jeffrey M Macdonald
- Joint Department of Biomedical Engineering, University of North Carolina , Chapel Hill, NC , USA
| | - Glenn Walker
- Joint Department of Biomedical Engineering, North Carolina State University , Raleigh, NC , USA
| | - Melvin E Andersen
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA; ScitoVation, LLC, Research Triangle Park, NC, USA
| | - Harvey J Clewell
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA; ScitoVation, LLC, Research Triangle Park, NC, USA
| | - Miyoung Yoon
- Institute for Chemical Safety Sciences, The Hamner Institutes for Health Sciences, Research Triangle Park, NC, USA; ScitoVation, LLC, Research Triangle Park, NC, USA
| |
Collapse
|
37
|
Lauschke VM, Hendriks DFG, Bell CC, Andersson TB, Ingelman-Sundberg M. Novel 3D Culture Systems for Studies of Human Liver Function and Assessments of the Hepatotoxicity of Drugs and Drug Candidates. Chem Res Toxicol 2016; 29:1936-1955. [DOI: 10.1021/acs.chemrestox.6b00150] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Volker M. Lauschke
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Delilah F. G. Hendriks
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Catherine C. Bell
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Tommy B. Andersson
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
- Cardiovascular
and Metabolic Diseases, Innovative Medicines and Early Development
Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Magnus Ingelman-Sundberg
- Section
of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| |
Collapse
|
38
|
Long TJ, Cosgrove PA, Dunn RT, Stolz DB, Hamadeh H, Afshari C, McBride H, Griffith LG. Modeling Therapeutic Antibody-Small Molecule Drug-Drug Interactions Using a Three-Dimensional Perfusable Human Liver Coculture Platform. Drug Metab Dispos 2016; 44:1940-1948. [PMID: 27621203 DOI: 10.1124/dmd.116.071456] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 09/08/2016] [Indexed: 12/16/2022] Open
Abstract
Traditional in vitro human liver cell culture models lose key hepatic functions such as metabolic activity during short-term culture. Advanced three-dimensional (3D) liver coculture platforms offer the potential for extended hepatocyte functionality and allow for the study of more complex biologic interactions, which can improve and refine human drug safety evaluations. Here, we use a perfusion flow 3D microreactor platform for the coculture of cryopreserved primary human hepatocytes and Kupffer cells to study the regulation of cytochrome P450 3A4 isoform (CYP3A4) activity by chronic interleukin 6 (IL-6)-mediated inflammation over 2 weeks. Hepatocyte cultures remained stable over 2 weeks, with consistent albumin production and basal IL-6 levels. Direct IL-6 stimulation that mimics an inflammatory state induced a dose-dependent suppression of CYP3A4 activity, an increase in C-reactive protein (CRP) secretion, and a decrease in shed soluble interleukin-6 receptor (IL-6R) levels, indicating expected hepatic IL-6 bioactivity. Tocilizumab, an anti-IL-6R monoclonal antibody used to treat rheumatoid arthritis, has been demonstrated clinically to impact small molecule drug pharmacokinetics by modulating cytochrome P450 enzyme activities, an effect not observed in traditional hepatic cultures. We have now recapitulated the clinical observation in a 3D bioreactor system. Tocilizumab was shown to desuppress CYP3A4 activity while reducing the CRP concentration after 72 hours in the continued presence of IL-6. This change in CYP3A4 activity decreased the half-life and area under the curve up to the last measurable concentration (AUClast) of the small molecule CYP3A4 substrate simvastatin hydroxy acid, measured before and after tocilizumab treatment. We conclude that next-generation in vitro liver culture platforms are well suited for these types of long-term treatment studies and show promise for improved drug safety assessment.
Collapse
Affiliation(s)
- Thomas J Long
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Patrick A Cosgrove
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Robert T Dunn
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Donna B Stolz
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Hisham Hamadeh
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Cynthia Afshari
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Helen McBride
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| | - Linda G Griffith
- Comparative Biology and Safety Science Laboratory, Amgen, Inc., Cambridge, Massachusetts (T.J.L.); Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (T.J.L., L.G.G.); Comparative Biology and Safety Science Laboratory, Amgen, Inc., Thousand Oaks, California (P.A.C., R.T.D., H.H., H.M., C.A.); Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, Massachusetts (L.G.G.); Center for Biologic Imaging, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.); Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania (D.B.S.)
| |
Collapse
|
39
|
Advances in high-resolution MS and hepatocyte models solve a long-standing metabolism challenge: the loratadine story. Bioanalysis 2016; 8:1645-62. [PMID: 27460981 DOI: 10.4155/bio-2016-0094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Loratadine (LOR, Claritin(®)) is a long-acting antihistamine used to treat allergic rhinitis. The major active human metabolite, desloratadine (DL, Clarinex(®)), is extensively metabolized to 3-hydroxydesloratadine (3-OH-DL) (M40) and subsequently glucuronidated before elimination. This study revealed the ability of a novel, long-term hepatocyte micropatterned co-culture (MPCC) model to generate in vivo metabolites. Metabolites were detected and characterized using non-targeted MS/MS(ALL) with SWATH™ acquisition by a UHPLC-Q-TOF system. Results & methodology: Human MPCCs extensively metabolized LOR and formed 3-OH-DL-glucuronide (M13). Cross-species comparisons revealed monkey- and rat-specific metabolites with gender-specific DL-pyridine-N-oxide formation in male rats. These results demonstrate a first for an in vitro hepatocyte model to generate circulating metabolites of LOR and detect species-specific differences. Early focus on human metabolites could have spared characterization of nonhuman metabolites in preclinical species.
Collapse
|