1
|
Mehta V, Karnam G, Madgula V. Liver-on-chips for drug discovery and development. Mater Today Bio 2024; 27:101143. [PMID: 39070097 PMCID: PMC11279310 DOI: 10.1016/j.mtbio.2024.101143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/07/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Recent FDA modernization act 2.0 has led to increasing industrial R&D investment in advanced in vitro 3D models such as organoids, spheroids, organ-on-chips, 3D bioprinting, and in silico approaches. Liver-related advanced in vitro models remain the prime area of interest, as liver plays a central role in drug clearance of compounds. Growing evidence indicates the importance of recapitulating the overall liver microenvironment to enhance hepatocyte maturity and culture longevity using liver-on-chips (LoC) in vitro. Hence, pharmaceutical industries have started exploring LoC assays in the two of the most challenging areas: accurate in vitro-in vivo extrapolation (IVIVE) of hepatic drug clearance and drug-induced liver injury. We examine the joint efforts of commercial chip manufacturers and pharmaceutical companies to present an up-to-date overview of the adoption of LoC technology in the drug discovery. Further, several roadblocks are identified to the rapid adoption of LoC assays in the current drug development framework. Finally, we discuss some of the underexplored application areas of LoC models, where conventional 2D hepatic models are deemed unsuitable. These include clearance prediction of metabolically stable compounds, immune-mediated drug-induced liver injury (DILI) predictions, bioavailability prediction with gut-liver systems, hepatic clearance prediction of drugs given during pregnancy, and dose adjustment studies in disease conditions. We conclude the review by discussing the importance of PBPK modeling with LoC, digital twins, and AI/ML integration with LoC.
Collapse
Affiliation(s)
- Viraj Mehta
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Guruswamy Karnam
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| | - Vamsi Madgula
- Organoid Technology Lab, DMPK Department, Sai Life Sciences, Hyderabad, 500078, India
| |
Collapse
|
2
|
Lee JM, Yoon JH, Maeng HJ, Kim YC. Physiologically Based Pharmacokinetic (PBPK) Modeling to Predict CYP3A-Mediated Drug Interaction between Saxagliptin and Nicardipine: Bridging Rat-to-Human Extrapolation. Pharmaceutics 2024; 16:280. [PMID: 38399334 PMCID: PMC10892660 DOI: 10.3390/pharmaceutics16020280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
The aim of this study was to predict the cytochrome P450 3A (CYP3A)-mediated drug-drug interactions (DDIs) between saxagliptin and nicardipine using a physiologically based pharmacokinetic (PBPK) model. Initially, in silico and in vitro parameters were gathered from experiments or the literature to construct PBPK models for each drug in rats. These models were integrated to predict the DDIs between saxagliptin, metabolized via CYP3A2, and nicardipine, exhibiting CYP3A inhibitory activity. The rat DDI PBPK model was completed by optimizing parameters using experimental rat plasma concentrations after co-administration of both drugs. Following co-administration in Sprague-Dawley rats, saxagliptin plasma concentration significantly increased, resulting in a 2.60-fold rise in AUC, accurately predicted by the rat PBPK model. Subsequently, the workflow of the rat PBPK model was applied to humans, creating a model capable of predicting DDIs between the two drugs in humans. Simulation from the human PBPK model indicated that nicardipine co-administration in humans resulted in a nearly unchanged AUC of saxagliptin, with an approximate 1.05-fold change, indicating no clinically significant changes and revealing a lack of direct translation of animal interaction results to humans. The animal-to-human PBPK model extrapolation used in this study could enhance the reliability of predicting drug interactions in clinical settings where DDI studies are challenging.
Collapse
Affiliation(s)
- Jeong-Min Lee
- Department of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Republic of Korea;
| | - Jin-Ha Yoon
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea;
| | - Han-Joo Maeng
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea;
| | - Yu Chul Kim
- Department of Digital Anti-Aging Healthcare, Inje University, Gimhae 50834, Republic of Korea;
- Department of Pharmaceutical Engineering, Inje University, Gimhae 50834, Republic of Korea
| |
Collapse
|
3
|
Macedo MH, Torras N, García-Díaz M, Barrias C, Sarmento B, Martínez E. The shape of our gut: Dissecting its impact on drug absorption in a 3D bioprinted intestinal model. BIOMATERIALS ADVANCES 2023; 153:213564. [PMID: 37482042 DOI: 10.1016/j.bioadv.2023.213564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
The small intestine is a complex organ with a characteristic architecture and a major site for drug and nutrient absorption. The three-dimensional (3D) topography organized in finger-like protrusions called villi increases surface area remarkably, granting a more efficient absorption process. The intestinal mucosa, where this process occurs, is a multilayered and multicell-type tissue barrier. In vitro intestinal models are routinely used to study different physiological and pathological processes in the gut, including compound absorption. Still, standard models are typically two-dimensional (2D) and represent only the epithelial barrier, lacking the cues offered by the 3D architecture and the stromal components present in vivo, often leading to inaccurate results. In this work, we studied the impact of the 3D architecture of the gut on drug transport using a bioprinted 3D model of the intestinal mucosa containing both the epithelial and the stromal compartments. Human intestinal fibroblasts were embedded in a previously optimized hydrogel bioink, and enterocytes and goblet cells were seeded on top to mimic the intestinal mucosa. The embedded fibroblasts thrived inside the hydrogel, remodeling the surrounding extracellular matrix. The epithelial cells fully covered the hydrogel scaffolds and formed a uniform cell layer with barrier properties close to in vivo. In particular, the villus-like model revealed overall increased permeability compared to a flat counterpart composed by the same hydrogel and cells. In addition, the efflux activity of the P-glycoprotein (P-gp) transporter was significantly reduced in the villus-like scaffold compared to a flat model, and the genetic expression of other drugs transporters was, in general, more relevant in the villus-like model. Globally, this study corroborates that the presence of the 3D architecture promotes a more physiological differentiation of the epithelial barrier, providing more accurate data on drug absorbance measurements.
Collapse
Affiliation(s)
- Maria Helena Macedo
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Núria Torras
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - María García-Díaz
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Cristina Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Rua Alfredo, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Elena Martínez
- IBEC - Institute for Bioengineering of Catalonia, BIST - The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain; CIBER-BBN - Consorcio Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Avenida Monforte de Lemos 3-5, 28029 Madrid, Spain; Electronics and Biomedical Engineering Department, Universitat de Barcelona, Martí I Franquès 1, 08028 Barcelona, Spain.
| |
Collapse
|
4
|
Jun J, Moon H, Yang S, Lee J, Baek J, Kim H, Cho H, Hwang K, Ahn S, Kim Y, Kim G, Kim H, Kwon H, Hah JM. Carbamate JNK3 Inhibitors Show Promise as Effective Treatments for Alzheimer's Disease: In Vivo Studies on Mouse Models. J Med Chem 2023; 66:6372-6390. [PMID: 37094094 DOI: 10.1021/acs.jmedchem.3c00393] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
We have been developing new inhibitors for c-Jun N-terminal kinase 3 (JNK3) as a potential treatment for Alzheimer's disease (AD). We identified potential JNK3 inhibitors through pharmacodynamic optimization studies, including benzimidazole compounds 2 and 3, but their unreliable pharmacokinetic properties led us to develop carbamate inhibitors 2h and 3h. In vitro studies validated carbamate inhibitors 2h and 3h as potent and highly selective JNK3 inhibitors with favorable pharmacokinetic profiles. Oral administration of 2h and 3h to both APP/PS1 and 3xTg AD mouse models improved cognitive function, indicating their potential as effective treatments for Alzheimer's disease. Carbamate JNK3 inhibitor 3h, in particular, restored cognitive function to near-normal levels in the 3xTg mice model of AD and led to pTau reduction in the hippocampal tissues of 3xTg-AD mice during in vivo behavioral evaluations. We intend to further develop these carbamate JNK3 inhibitors in preclinical studies as a potential first-in-class treatment for AD.
Collapse
Affiliation(s)
- Joonhong Jun
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Hyungwoo Moon
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Songyi Yang
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Junghun Lee
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Jihyun Baek
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Hyejin Kim
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Hyunwook Cho
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| | - Kyungrim Hwang
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - Soyeon Ahn
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - Yuro Kim
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - Gibeom Kim
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - HyunTae Kim
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - Hoseok Kwon
- Research Center, Samjin Pharm. Co. Ltd. 90, Magokjungang 10-ro, Gangseo-gu, Seoul 07794, Republic of Korea
| | - Jung-Mi Hah
- Department of Pharmacy & Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-ro, Sangnok-gu, Ansan, Kyeonggi-do 15588, Republic of Korea
| |
Collapse
|
5
|
Jun J, Yang S, Lee J, Moon H, Kim J, Jung H, Im D, Oh Y, Jang M, Cho H, Baek J, Kim H, Kang D, Bae H, Tak C, Hwang K, Kwon H, Kim H, Hah JM. Discovery of novel imidazole chemotypes as isoform-selective JNK3 inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2023; 245:114894. [DOI: 10.1016/j.ejmech.2022.114894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
|
6
|
Weller A, Hansen MB, Marie R, Hundahl AC, Hempel C, Kempen PJ, Frandsen HL, Parhamifar L, Larsen JB, Andresen TL. Quantifying the transport of biologics across intestinal barrier models in real-time by fluorescent imaging. Front Bioeng Biotechnol 2022; 10:965200. [PMID: 36159696 PMCID: PMC9500407 DOI: 10.3389/fbioe.2022.965200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Unsuccessful clinical translation of orally delivered biological drugs remains a challenge in pharmaceutical development and has been linked to insufficient mechanistic understanding of intestinal drug transport. Live cell imaging could provide such mechanistic insights by directly tracking drug transport across intestinal barriers at subcellular resolution, however traditional intestinal in vitro models are not compatible with the necessary live cell imaging modalities. Here, we employed a novel microfluidic platform to develop an in vitro intestinal epithelial barrier compatible with advanced widefield- and confocal microscopy. We established a quantitative, multiplexed and high-temporal resolution imaging assay for investigating the cellular uptake and cross-barrier transport of biologics while simultaneously monitoring barrier integrity. As a proof-of-principle, we use the generic model to monitor the transport of co-administrated cell penetrating peptide (TAT) and insulin. We show that while TAT displayed a concentration dependent difference in its transport mechanism and efficiency, insulin displayed cellular internalization, but was restricted from transport across the barrier. This illustrates how such a sophisticated imaging based barrier model can facilitate mechanistic studies of drug transport across intestinal barriers and aid in vivo and clinical translation in drug development.
Collapse
Affiliation(s)
- Arjen Weller
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Morten B. Hansen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Rodolphe Marie
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Adam C. Hundahl
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Casper Hempel
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Paul J. Kempen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- The National Centre for Nano Fabrication and Characterization, DTU Nanolab, Technical University of Denmark, Lyngby, Denmark
| | - Henrik L. Frandsen
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Ladan Parhamifar
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Jannik B. Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- *Correspondence: Jannik B. Larsen, ; Thomas L. Andresen,
| | - Thomas L. Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- *Correspondence: Jannik B. Larsen, ; Thomas L. Andresen,
| |
Collapse
|
7
|
The Caco-2 Model: Modifications and enhancements to improve efficiency and predictive performance. Int J Pharm 2022; 624:122004. [PMID: 35820514 DOI: 10.1016/j.ijpharm.2022.122004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/28/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022]
Abstract
The Caco-2 cell model has been widely used to assess the permeability of drug candidates. It has provided a high throughput in vitro platform, functionally resembling the enterocytes. Since the oral route is the most preferred for drug administration, the Caco-2 cell model acts as a very important tool to elucidate the oral "druggability" of a molecule by providing a fairly reliable estimate of its permeability through the intestinal membrane. Despite its shortcomings (the lack of a mucus layer, long cultivation period, inter-lab variability, and differences in expression of enzymes, transporters, and tight junction complexes) it remains heavily used due to its reliability, predictive performance, and wide acceptance. Various modifications have been made: co-culturing with other intestinal cells, applying biosimilar mucus, reducing culturing time, combining Caco-2 monolayer with the dissolution apparatus, enhancing protein expression, and redesigning the sampling apparatus. These modifications are intended to overcome some of the shortcomings of the Caco-2 model in order to make its use easier, quicker, economical, and more representative of the intestine. The aim of this review is to discuss such modifications to enhance this model's utility, predictive performance, and reproducibility.
Collapse
|
8
|
Klepach A, Tran H, Ahmad Mohammed F, ElSayed ME. Characterization and impact of peptide physicochemical properties on oral and subcutaneous delivery. Adv Drug Deliv Rev 2022; 186:114322. [PMID: 35526665 DOI: 10.1016/j.addr.2022.114322] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/21/2022] [Accepted: 05/02/2022] [Indexed: 11/25/2022]
Abstract
Peptides, an emerging modality within the biopharmaceutical industry, are often delivered subcutaneously with evolving prospects on oral delivery. Barrier biology within the subcutis or gastrointestinal tract is a significant challenge in limiting absorption or otherwise disrupting peptide disposition. Aspects of peptide pharmacokinetic performance and ADME can be mitigated with careful molecular design that tailors for properties such as effective size, hydrophobicity, net charge, proteolytic stability, and albumin binding. In this review, we endeavor to highlight effective techniques in qualifying physicochemical properties of peptides and discuss advancements of in vitro models of subcutaneous and oral delivery. Additionally, we will delineate empirical findings around the relationship of these physicochemical properties and in vivo (animal or human) impact. We conclude that robust peptide characterization methods and in vitro techniques with demonstrated correlations to in vivo data are key routines to incorporate in the drug discovery and development to improve the probability of technical and commercial success of peptide therapeutics.
Collapse
|
9
|
Park J, Heo YJ, Kwon S. Interaction Between Hepatocytes and Proximal Tubular Epithelial Cells in Hypoxia-induced Lipotoxicity. BIOTECHNOL BIOPROC E 2021. [DOI: 10.1007/s12257-021-0137-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
10
|
All layers matter: Innovative three-dimensional epithelium-stroma-endothelium intestinal model for reliable permeability outcomes. J Control Release 2021; 341:414-430. [PMID: 34871636 DOI: 10.1016/j.jconrel.2021.11.048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 12/15/2022]
Abstract
Drug development is an ever-growing field, increasingly requesting reliable in vitro tools to speed up early screening phases, reducing the need for animal experiments. In oral delivery, understanding the absorption pattern of a new drug in the small intestine is paramount. Classical two-dimensional (2D) in vitro models are generally too simplistic and do not accurately represent native tissues. The main goal of this work was to develop an advanced three-dimensional (3D) in vitro intestinal model to test absorption in a more reliable manner, by better mimicking the native environment. The 3D model is composed of a collagen-based stromal layer with embedded fibroblasts mimicking the intestinal lamina propria and providing support for the epithelium, composed of enterocytes and mucus-secreting cells. An endothelial layer, surrogating the absorptive capillary network, is also present. The cellular crosstalk between the different cells present in the model is unveiled, disclosing key players, namely those involved in the contraction of collagen by fibroblasts. The developed 3D model presents lower levels of P-glycoprotein (P-gp) and Multidrug Resistance Protein 2 (MRP2) efflux transporters, which are normally overexpressed in traditional Caco-2 models, and are paramount in the absorption of many compounds. This, allied with transepithelial electrical resistance (TEER) values closer to physiological ranges, leads to improved and more reliable permeability outcomes, which are observed when comparing our results with in vivo data.
Collapse
|
11
|
Hagiwara Y, Kumagai H, Ouwerkerk N, Gijzen L, Annida R, Bokkers M, van Vught R, Yoshinari K, Katakawa Y, Motonaga K, Tajiri T. A Novel In Vitro Membrane Permeability Methodology Using Three-dimensional Caco-2 Tubules in a Microphysiological System Which Better Mimics In Vivo Physiological Conditions. J Pharm Sci 2021; 111:214-224. [PMID: 34838780 DOI: 10.1016/j.xphs.2021.11.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/23/2021] [Accepted: 11/23/2021] [Indexed: 01/27/2023]
Abstract
The aim of this study was to develop an in vitro drug permeability methodology which mimics the gastrointestinal environment more accurately than conventional 2D methodologies through a three-dimensional (3D) Caco-2 tubules using a microphysiological system. Such a system offers significant advantages, including accelerated cellular polarization and more accurate mimicry of the in vivo environment. This methodology was confirmed by measuring the permeability of propranolol as a model compound, and subsequently applied to those of solifenacin and bile acids for a comprehensive understanding of permeability for the drug product in the human gastrointestinal tract. To protect the Caco-2 tubules from bile acid toxicity, a mucus layer was applied on the surface of Caco-2 tubules and it enables to use simulated intestinal fluid. The assessment using propranolol reproduced results equivalent to those obtained from conventional methodology, while that using solifenacin indicated fluctuations in the permeability of solifenacin due to various factors, including interaction with bile acids. We therefore suggest that this model will serve as an alternative testing system for measuring drug absorption in an environment closely resembling that of the human gastrointestinal tract.
Collapse
Affiliation(s)
- Yuki Hagiwara
- Analytical Research Laboratories, Astellas Pharma Inc., Yaizu, Shizuoka 425-0072, Japan
| | - Harumi Kumagai
- Analytical Research Laboratories, Astellas Pharma Inc., Yaizu, Shizuoka 425-0072, Japan
| | - Niels Ouwerkerk
- European Analytical Research Laboratories, Astellas Pharma Europe B.V., Leiden 2333 BE, the Netherlands
| | - Linda Gijzen
- Mimetas B.V., Oegstgeest 2342 DH, the Netherlands
| | | | | | | | - Kouichi Yoshinari
- Laboratory of Molecular Toxicology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Yoshifumi Katakawa
- Analytical Research Laboratories, Astellas Pharma Inc., Yaizu, Shizuoka 425-0072, Japan
| | - Kei Motonaga
- Analytical Research Laboratories, Astellas Pharma Inc., Yaizu, Shizuoka 425-0072, Japan
| | - Tomokazu Tajiri
- Pharmaceutical Science and Technology Laboratories, Astellas Pharma Inc., Tsukuba, Ibaraki 300-2698, Japan.
| |
Collapse
|
12
|
Larder CE, Iskandar MM, Kubow S. Assessment of Bioavailability after In Vitro Digestion and First Pass Metabolism of Bioactive Peptides from Collagen Hydrolysates. Curr Issues Mol Biol 2021; 43:1592-1605. [PMID: 34698092 PMCID: PMC8928955 DOI: 10.3390/cimb43030113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/07/2021] [Accepted: 10/09/2021] [Indexed: 12/21/2022] Open
Abstract
Collagen hydrolysates (CHs) are composed of bioactive peptides (BAPs), which possess health enhancing properties. There is a knowledge gap regarding the bioavailability of these BAPs that involves intestinal transport and hepatic first pass effects. A simulated gastrointestinal model was used to generate digesta from two CHs (CH-GL and CH-OPT), which were applied to a novel transwell co-culture of human intestinal epithelium cell line-6 (HIEC-6) and hepatic (HepG2) cells to simulate in vivo conditions of absorption and first pass metabolism. Peptide transport, hepatic first pass effects, and bioavailability were determined by measuring BAPs (Gly-Pro, Hyp-Gly, Ala-Hyp, Pro-Hyp, Gly-Pro-Hyp) using an innovative capillary electrophoresis method. All peptides were transported across the intestinal cell layer to varying degrees with both CHs; however, Gly-Pro-Hyp was transported only with CH-GL, but not CH-OPT. Notable hepatic production was observed for Ala-Hyp with both CH treatments, and for Pro-Hyp and Gly-Pro with CH-GL only. All peptides were bioavailable (>10%), except for Gly-Pro-Hyp after CH-OPT. Overall, a high degree of transport and hepatic first pass effects on CH-derived BAPs were observed. Further research is needed to explore the hepatic mechanisms related to the production of BAPs and the bifunctional effects of the bioavailable BAPs noted in this study.
Collapse
|
13
|
Fedi A, Vitale C, Ponschin G, Ayehunie S, Fato M, Scaglione S. In vitro models replicating the human intestinal epithelium for absorption and metabolism studies: A systematic review. J Control Release 2021; 335:247-268. [PMID: 34033859 DOI: 10.1016/j.jconrel.2021.05.028] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/18/2021] [Accepted: 05/20/2021] [Indexed: 12/17/2022]
Abstract
Absorption, distribution, metabolism and excretion (ADME) studies represent a fundamental step in the early stages of drug discovery. In particular, the absorption of orally administered drugs, which occurs at the intestinal level, has gained attention since poor oral bioavailability often led to failures for new drug approval. In this context, several in vitro preclinical models have been recently developed and optimized to better resemble human physiology in the lab and serve as an animal alternative to accomplish the 3Rs principles. However, numerous models are ineffective in recapitulating the key features of the human small intestine epithelium and lack of prediction potential for drug absorption and metabolism during the preclinical stage. In this review, we provide an overview of in vitro models aimed at mimicking the intestinal barrier for pharmaceutical screening. After briefly describing how the human small intestine works, we present i) conventional 2D synthetic and cell-based systems, ii) 3D models replicating the main features of the intestinal architecture, iii) micro-physiological systems (MPSs) reproducing the dynamic stimuli to which cells are exposed in the native microenvironment. In this review, we will highlight the benefits and drawbacks of the leading intestinal models used for drug absorption and metabolism studies.
Collapse
Affiliation(s)
- Arianna Fedi
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy; National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Chiara Vitale
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Giulia Ponschin
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy
| | | | - Marco Fato
- Department of Computer Science, Bioengineering, Robotics and Systems Engineering, University of Genoa, 16126 Genoa, Italy; National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy
| | - Silvia Scaglione
- National Research Council of Italy, Institute of Electronics, Computer and Telecommunications (IEIIT) Institute, 16149 Genoa, Italy.
| |
Collapse
|
14
|
Kulthong K, Hooiveld GJEJ, Duivenvoorde L, Miro Estruch I, Marin V, van der Zande M, Bouwmeester H. Transcriptome comparisons of in vitro intestinal epithelia grown under static and microfluidic gut-on-chip conditions with in vivo human epithelia. Sci Rep 2021; 11:3234. [PMID: 33547413 PMCID: PMC7864925 DOI: 10.1038/s41598-021-82853-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/22/2021] [Indexed: 02/06/2023] Open
Abstract
Gut-on-chip devices enable exposure of cells to a continuous flow of culture medium, inducing shear stresses and could thus better recapitulate the in vivo human intestinal environment in an in vitro epithelial model compared to static culture methods. We aimed to study if dynamic culture conditions affect the gene expression of Caco-2 cells cultured statically or dynamically in a gut-on-chip device and how these gene expression patterns compared to that of intestinal segments in vivo. For this we applied whole genome transcriptomics. Dynamic culture conditions led to a total of 5927 differentially expressed genes (3280 upregulated and 2647 downregulated genes) compared to static culture conditions. Gene set enrichment analysis revealed upregulated pathways associated with the immune system, signal transduction and cell growth and death, and downregulated pathways associated with drug metabolism, compound digestion and absorption under dynamic culture conditions. Comparison of the in vitro gene expression data with transcriptome profiles of human in vivo duodenum, jejunum, ileum and colon tissue samples showed similarities in gene expression profiles with intestinal segments. It is concluded that both the static and the dynamic gut-on-chip model are suitable to study human intestinal epithelial responses as an alternative for animal models.
Collapse
Affiliation(s)
- Kornphimol Kulthong
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands.
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands.
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency, Pathum Thani, 12120, Thailand.
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Loes Duivenvoorde
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Ignacio Miro Estruch
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands
| | - Victor Marin
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Meike van der Zande
- Wageningen Food Safety Research, P.O. Box 230, 6700 AE, Wageningen, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, P.O. box 8000, 6700 EA, Wageningen, The Netherlands.
| |
Collapse
|
15
|
Abstract
Accurate estimation of in vivo clearance in human is pivotal to determine the dose and dosing regimen for drug development. In vitro-in vivo extrapolation (IVIVE) has been performed to predict drug clearance using empirical and physiological scalars. Multiple in vitro systems and mathematical modeling techniques have been employed to estimate in vivo clearance. The models for predicting clearance have significantly improved and have evolved to become more complex by integrating multiple processes such as drug metabolism and transport as well as passive diffusion. This chapter covers the use of conventional as well as recently developed methods to predict metabolic and transporter-mediated clearance along with the advantages and disadvantages of using these methods and the associated experimental considerations. The general approaches to improve IVIVE by use of appropriate scalars, incorporation of extrahepatic metabolism and transport and application of physiologically based pharmacokinetic (PBPK) models with proteomics data are also discussed. The chapter also provides an overview of the advantages of using such dynamic mechanistic models over static models for clearance predictions to improve IVIVE.
Collapse
|
16
|
Docci L, Umehara K, Krähenbühl S, Fowler S, Parrott N. Construction and Verification of Physiologically Based Pharmacokinetic Models for Four Drugs Majorly Cleared by Glucuronidation: Lorazepam, Oxazepam, Naloxone, and Zidovudine. AAPS JOURNAL 2020; 22:128. [DOI: 10.1208/s12248-020-00513-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023]
|
17
|
Microfluidic chip for culturing intestinal epithelial cell layers: Characterization and comparison of drug transport between dynamic and static models. Toxicol In Vitro 2020; 65:104815. [DOI: 10.1016/j.tiv.2020.104815] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/28/2020] [Indexed: 12/29/2022]
|
18
|
Leth Jepsen M, Willumsen A, Mazzoni C, Boisen A, Hagner Nielsen L, Dufva M. 3D Printed Stackable Titer Plate Inserts Supporting Three Interconnected Tissue Models for Drug Transport Studies. ACTA ACUST UNITED AC 2020; 4:e1900289. [DOI: 10.1002/adbi.201900289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/17/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Morten Leth Jepsen
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| | - Andreas Willumsen
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| | - Chiara Mazzoni
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| | - Anja Boisen
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| | - Line Hagner Nielsen
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| | - Martin Dufva
- The Danish National Research Foundation and Villum Foundation’s Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN)Department of Health TechnologyTechnical University of Denmark Ørsteds Plads 345C Kgs. Lyngby 2800 Denmark
| |
Collapse
|
19
|
Corazza FG, Ernesto JV, Nambu FA, de Carvalho LR, Leite-Silva VR, Varca GH, Calixto LA, Vieira DP, Andréo-Filho N, Lopes PS. Papain-cyclodextrin complexes as an intestinal permeation enhancer: Permeability and in vitro safety evaluation. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101413] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
20
|
Prantil-Baun R, Novak R, Das D, Somayaji MR, Przekwas A, Ingber DE. Physiologically Based Pharmacokinetic and Pharmacodynamic Analysis Enabled by Microfluidically Linked Organs-on-Chips. Annu Rev Pharmacol Toxicol 2019; 58:37-64. [PMID: 29309256 DOI: 10.1146/annurev-pharmtox-010716-104748] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling and simulation approaches are beginning to be integrated into drug development and approval processes because they enable key pharmacokinetic (PK) parameters to be predicted from in vitro data. However, these approaches are hampered by many limitations, including an inability to incorporate organ-specific differentials in drug clearance, distribution, and absorption that result from differences in cell uptake, transport, and metabolism. Moreover, such approaches are generally unable to provide insight into pharmacodynamic (PD) parameters. Recent development of microfluidic Organ-on-a-Chip (Organ Chip) cell culture devices that recapitulate tissue-tissue interfaces, vascular perfusion, and organ-level functionality offer the ability to overcome these limitations when multiple Organ Chips are linked via their endothelium-lined vascular channels. Here, we discuss successes and challenges in the use of existing culture models and vascularized Organ Chips for PBPK and PD modeling of human drug responses, as well as in vitro to in vivo extrapolation (IVIVE) of these results, and how these approaches might advance drug development and regulatory review processes in the future.
Collapse
Affiliation(s)
- Rachelle Prantil-Baun
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Richard Novak
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA;
| | - Debarun Das
- CFD Research Corporation, Huntsville, Alabama 35806, USA
| | | | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, USA; .,Vascular Biology Program and Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
21
|
The In Vivo and In Vitro Toxicokinetics of Citreoviridin Extracted from Penicillium citreonigrum. Toxins (Basel) 2019; 11:toxins11060360. [PMID: 31226823 PMCID: PMC6628624 DOI: 10.3390/toxins11060360] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 11/17/2022] Open
Abstract
Citreoviridin (CTVD), a mycotoxin called yellow rice toxin, is reported to be related to acute cardiac beriberi; however, its toxicokinetics remain unclear. The present study elucidated the toxicokinetics through in vivo experiments in swine and predicted the human toxicokinetics by comparing the findings to those from in vitro experiments. In vivo experiments revealed the high bioavailability of CTVD (116.4%) in swine. An intestinal permeability study using Caco-2 cells to estimate the toxicokinetics in humans showed that CTVD has a high permeability coefficient. When CTVD was incubated with hepatic S9 fraction from swine and humans, hydroxylation and methylation, desaturation, and dihydroxylation derivatives were produced as the predominant metabolites. The levels of these products produced using human S9 were higher than those obtained swine S9, while CTVD glucuronide was produced slowly in human S9 in comparison to swine S9. Furthermore, the elimination of CTVD by human S9 was significantly more rapid in comparison to that by swine S9. These results suggest that CTVD is easily absorbed in swine and that it remains in the body where it is slowly metabolized. In contrast, the absorption of CTVD in humans would be the same as that in swine, although its elimination would be faster.
Collapse
|
22
|
Pham-The H, Cabrera-Pérez MÁ, Nam NH, Castillo-Garit JA, Rasulev B, Le-Thi-Thu H, Casañola-Martin GM. In Silico Assessment of ADME Properties: Advances in Caco-2 Cell Monolayer Permeability Modeling. Curr Top Med Chem 2019; 18:2209-2229. [PMID: 30499410 DOI: 10.2174/1568026619666181130140350] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/16/2018] [Accepted: 11/19/2018] [Indexed: 11/22/2022]
Abstract
One of the main goals of in silico Caco-2 cell permeability models is to identify those drug substances with high intestinal absorption in human (HIA). For more than a decade, several in silico Caco-2 models have been made, applying a wide range of modeling techniques; nevertheless, their capacity for intestinal absorption extrapolation is still doubtful. There are three main problems related to the modest capacity of obtained models, including the existence of inter- and/or intra-laboratory variability of recollected data, the influence of the metabolism mechanism, and the inconsistent in vitro-in vivo correlation (IVIVC) of Caco-2 cell permeability. This review paper intends to sum up the recent advances and limitations of current modeling approaches, and revealed some possible solutions to improve the applicability of in silico Caco-2 permeability models for absorption property profiling, taking into account the above-mentioned issues.
Collapse
Affiliation(s)
- Hai Pham-The
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Miguel Á Cabrera-Pérez
- Unit of Modeling and Experimental Biopharmaceutics, Chemical Bioactive Center, Central University of Las Villas, Santa Clara, 54830, Villa Clara, Cuba.,Department of Engineering, Area of Pharmacy and Pharmaceutical Technology, Miguel Hernández University, 03550 Sant Juan d'Alacant, Alicante, Spain
| | - Nguyen-Hai Nam
- Hanoi University of Pharmacy, 13-15 Le Thanh Tong, Hanoi, Vietnam
| | - Juan A Castillo-Garit
- Unidad de Toxicologia Experimental, Universidad de Ciencias Medicas "Dr. Serafín Ruiz de Zarate Ruiz" de Villa Clara, Santa Clara, 50200, Villa Clara, Cuba
| | - Bakhtiyor Rasulev
- Department of Coatings and Polymer Materials, North Dakota State University, Fargo, ND, 58102, United States
| | - Huong Le-Thi-Thu
- School of Medicine and Pharmacy, Vietnam National University, 144 Xuan Thuy, Hanoi, Vietnam
| | - Gerardo M Casañola-Martin
- Department of Coatings and Polymer Materials, North Dakota State University, Fargo, ND, 58102, United States
| |
Collapse
|
23
|
Yamada M, Mendell J, Takakusa H, Shimizu T, Ando O. Pharmacokinetics, Metabolism, and Excretion of [ 14C]Esaxerenone, a Novel Mineralocorticoid Receptor Blocker in Humans. Drug Metab Dispos 2018; 47:340-349. [PMID: 30541878 DOI: 10.1124/dmd.118.084897] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/05/2018] [Indexed: 11/22/2022] Open
Abstract
Esaxerenone (CS-3150) is a novel, nonsteroidal, selective mineralocorticoid receptor blocker. The absorption, metabolism, distribution, and excretion of esaxerenone were assessed in in vitro studies and in a clinical study, where [14C]esaxerenone (150 μCi/20 mg) was administered orally to six healthy male subjects. The plasma concentrations of esaxerenone and its metabolites (M4, M11, and M1) were measured using liquid chromatography-tandem mass spectrometry. The recovery of radioactivity was 92.5%, with 38.5% and 54.0% excreted in the urine and feces, respectively. The half-life of radioactivity in blood and plasma was approximately 30 hours, similar to that of the unchanged form in plasma. The blood-to-plasma ratio was 0.628, demonstrating low partitioning to blood components. In plasma, esaxerenone was the most abundant moiety (40.8%), followed by O-glucuronide (21.4%; M4), acyl-glucuronide of amide-bond hydrolysate (8.0%; M11), and the deshydroxyethyl form (1.7%; M1). In vitro studies showed that esaxerenone was a substrate of CYP3A and multiple UDP-glucuronosyltransferase isoforms. Oxidation contributed approximately 30% to its clearance, as indicated by the excretion ratio of oxidized metabolites into urine and feces. Caco-2 studies showed that esaxerenone was a substrate of P-glycoprotein and breast cancer resistance protein; however, the excretion ratios of the unchanged form in the feces and urine were 18.7% and 1.6%, respectively, indicating that these transporters were not important for the absorption and elimination of esaxerenone. Low urinary excretion of esaxerenone suggested that the plasma exposure of esaxerenone was not affected by renal dysfunction. Multiple elimination pathways including oxidation, glucuronidation, and hydrolysis, and the low contribution of transporters, indicated limited drug-drug interaction potential.
Collapse
Affiliation(s)
- Makiko Yamada
- Drug Metabolism and Pharmacokinetics Research Laboratories (M.Y., H.T., O.A.) and Clinical Pharmacology Department (T.S.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; and Daiichi Sankyo Pharma Development, Basking Ridge, New Jersey (J.M.)
| | - Jeanne Mendell
- Drug Metabolism and Pharmacokinetics Research Laboratories (M.Y., H.T., O.A.) and Clinical Pharmacology Department (T.S.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; and Daiichi Sankyo Pharma Development, Basking Ridge, New Jersey (J.M.)
| | - Hideo Takakusa
- Drug Metabolism and Pharmacokinetics Research Laboratories (M.Y., H.T., O.A.) and Clinical Pharmacology Department (T.S.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; and Daiichi Sankyo Pharma Development, Basking Ridge, New Jersey (J.M.)
| | - Takako Shimizu
- Drug Metabolism and Pharmacokinetics Research Laboratories (M.Y., H.T., O.A.) and Clinical Pharmacology Department (T.S.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; and Daiichi Sankyo Pharma Development, Basking Ridge, New Jersey (J.M.)
| | - Osamu Ando
- Drug Metabolism and Pharmacokinetics Research Laboratories (M.Y., H.T., O.A.) and Clinical Pharmacology Department (T.S.), Daiichi Sankyo Co., Ltd., Tokyo, Japan; and Daiichi Sankyo Pharma Development, Basking Ridge, New Jersey (J.M.)
| |
Collapse
|
24
|
Lindmark B, Lundahl A, Kanebratt KP, Andersson TB, Isin EM. Human hepatocytes and cytochrome P450-selective inhibitors predict variability in human drug exposure more accurately than human recombinant P450s. Br J Pharmacol 2018; 175:2116-2129. [PMID: 29574682 PMCID: PMC5980217 DOI: 10.1111/bph.14203] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 02/27/2018] [Accepted: 03/02/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Drugs metabolically eliminated by several enzymes are less vulnerable to variable compound exposure in patients due to drug-drug interactions (DDI) or if a polymorphic enzyme is involved in their elimination. Therefore, it is vital in drug discovery to accurately and efficiently estimate and optimize the metabolic elimination profile. EXPERIMENTAL APPROACH CYP3A and/or CYP2D6 substrates with well described variability in vivo in humans due to CYP3A DDI and CYP2D6 polymorphism were selected for assessment of fraction metabolized by each enzyme (fmCYP ) in two in vitro systems: (i) human recombinant P450s (hrP450s) and (ii) human hepatocytes combined with selective P450 inhibitors. Increases in compound exposure in poor versus extensive CYP2D6 metabolizers and by the strong CYP3A inhibitor ketoconazole were mathematically modelled and predicted changes in exposure were compared with in vivo data. KEY RESULTS Predicted changes in exposure were within twofold of reported in vivo values using fmCYP estimated in human hepatocytes and there was a strong linear correlation between predicted and observed changes in exposure (r2 = 0.83 for CYP3A, r2 = 0.82 for CYP2D6). Predictions using fmCYP in hrP450s were not as accurate (r2 = 0.55 for CYP3A, r2 = 0.20 for CYP2D6). CONCLUSIONS AND IMPLICATIONS The results suggest that variability in human drug exposure due to DDI and enzyme polymorphism can be accurately predicted using fmCYP from human hepatocytes and CYP-selective inhibitors. This approach can be efficiently applied in drug discovery to aid optimization of candidate drugs with a favourable metabolic elimination profile and limited variability in patients.
Collapse
Affiliation(s)
- Bo Lindmark
- Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| | - Anna Lundahl
- Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| | - Kajsa P Kanebratt
- Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| | - Tommy B Andersson
- Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| | - Emre M Isin
- Cardiovascular, Renal and Metabolism, Innovative Medicines and Early Development Biotech UnitAstraZenecaGothenburgSweden
| |
Collapse
|
25
|
Yellepeddi VK, Zhudeva MY, Movahedi F, Vo A, Phan J, Kirsh RD, Rawlins DB, Talbot JN. Biopharmaceutical Characterization and Oral Efficacy of a New Rapid Acting Antidepressant Ro 25-6981. J Pharm Sci 2018; 107:2472-2478. [PMID: 29800545 DOI: 10.1016/j.xphs.2018.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/14/2018] [Accepted: 05/16/2018] [Indexed: 12/25/2022]
Abstract
Ro 25-6981 is a highly potent and selective blocker of N-methyl-d-aspartate receptors that has been shown to possess both rapid and sustained antidepressant activity. In the present study, we report the biopharmaceutical characterization of Ro 25-6981 by evaluating gastrointestinal stability, transepithelial permeability, stability in human liver microsomes, and in silico metabolic prediction. Moreover, in vivo efficacy of Ro 25-6981 after oral administration was evaluated in animal models of depression. When mixed with 5 different simulated gastrointestinal fluids, no loss of parent compound was observed after 6 h, indicating compound stability in the gastrointestinal environment. At the tested concentrations, Ro 25-6981 was shown to have transepithelial permeability with apparent permeability (Papp) values comparable to highly permeable drugs. Ro 25-6981 was metabolized within 30 min in human liver microsomes, and the metabolic prediction data showed glucuronidation and sulfation as potential metabolic pathways. The in vivo efficacy data suggested that Ro 25-6981, when administered orally at 30 mg/kg, exhibits antidepressant-like activity following oral administration with efficacy comparable to traditional antidepressants that is both dose- and time-dependent. Overall, due to optimal gastrointestinal stability, oral permeability, and oral efficacy, Ro 25-6981 can be a potential therapeutic option for the treatment of depression.
Collapse
Affiliation(s)
- Venkata K Yellepeddi
- Division of Clinical Pharmacology, Department of Pediatrics, University of Utah, Salt Lake City, Utah 84112; Utah Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah 84112; Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah 84112.
| | - Maryia Y Zhudeva
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095
| | - Fereshteh Movahedi
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095
| | - Annh Vo
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095
| | - Julie Phan
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095
| | - Robert D Kirsh
- Comparative Medicine Unit, Roseman University of Health Sciences, South Jordan, Utah 84095
| | - David B Rawlins
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095; Research Center on Substance Abuse and Depression, Roseman University of Health Sciences, Henderson, Nevada 89014
| | - Jeffery N Talbot
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, Utah 84095; Research Center on Substance Abuse and Depression, Roseman University of Health Sciences, Henderson, Nevada 89014
| |
Collapse
|
26
|
Sadeghi Ekbatan S, Iskandar MM, Sleno L, Sabally K, Khairallah J, Prakash S, Kubow S. Absorption and Metabolism of Phenolics from Digests of Polyphenol-Rich Potato Extracts Using the Caco-2/HepG2 Co-Culture System. Foods 2018; 7:foods7010008. [PMID: 29329242 PMCID: PMC5789271 DOI: 10.3390/foods7010008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/23/2017] [Accepted: 01/09/2018] [Indexed: 01/15/2023] Open
Abstract
The bioactivity of dietary polyphenols depends upon gastrointestinal and hepatic metabolism of secondary microbial phenolic metabolites generated via colonic microbiota-mediated biotransformation. A polyphenol-rich potato extract (PRPE) containing chlorogenic, caffeic, and ferulic acids and rutin was digested in a dynamic multi-reactor gastrointestinal simulator of the human intestinal microbial ecosystem (GI model). Simulated digestion showed extensive degradation of the parent compounds and the generation of microbial phenolic metabolites. To characterize the transport and metabolism of microbial phenolic metabolites following digestion, a co-culture of intestinal Caco-2 and hepatic HepG2 cells was exposed to the PRPE-derived digests obtained from the colonic vessels. Following a 2 h incubation of the digesta with the Caco-2/HepG2 co-cultures, approximately 10–15% of ferulic, dihydrocaffeic, and dihydroferulic acids and 3–5% of 3-hydroxybenzoic, 3-hydroxyphenylpropionic, and coumaric acids were observed in the basolateral side, whereas 3-hydroxyphenylacetic acid, phenylpropanoic acid, and cinnamic acid were not detected. Subsequent HepG2 cellular metabolism led to major increases in ferulic, dihydrocaffeic, 3-hydroxyphenylpropionic, and coumaric acids ranging from 160–370%. These findings highlight the importance of hepatic metabolism towards the generation of secondary metabolites of polyphenols despite low selective Caco-2 cellular uptake of microbial phenolic metabolites.
Collapse
Affiliation(s)
- Shima Sadeghi Ekbatan
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Michele M Iskandar
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Lekha Sleno
- Chemistry Department, Université du Québec à Montréal, 2101 Jeanne-Mance, Montreal, QC H2X2J6, Canada.
| | - Kebba Sabally
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Joelle Khairallah
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| | - Satya Prakash
- Department of Biomedical Engineering, Duff Medical Building, 3775 Rue University, Montreal, QC H3A2B4, Canada.
| | - Stan Kubow
- School of Human Nutrition, McGill University, 21111 Lakeshore, Ste. Anne de Bellevue, Montreal, QC H9X3V9, Canada.
| |
Collapse
|
27
|
González-Arias CA, Marín S, Rojas-García AE, Sanchis V, Ramos AJ. UPLC-MS/MS analysis of ochratoxin A metabolites produced by Caco-2 and HepG2 cells in a co-culture system. Food Chem Toxicol 2017; 109:333-340. [PMID: 28888735 DOI: 10.1016/j.fct.2017.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/22/2017] [Accepted: 09/05/2017] [Indexed: 12/21/2022]
Abstract
Ochatoxin A (OTA) is one of the most important mycotoxins based on its toxicity. The oral route is the main gateway of entry of OTA into the human body, and specialized epithelial cells constitute the first barrier. The present study investigated the in vitro cytotoxic effect of OTA (5, 15 and 45 μM) and production of OTA metabolities in Caco-2 and HepG2 cells using a co-culture Transwell System to mimic the passage through the intestinal epithelium and hepatic metabolism. The results derived from MTS cell viability assays and transepithelial electrical resistance measurements showed that OTA was slightly cytotoxic at the lowest concentration at 3 h, but significant toxicity was observed at all concentrations at 24 h. OTA metabolites generated in this co-culture were ochratoxin B (OTB), OTA methyl ester, OTA ethyl ester and the OTA glutathione conjugate (OTA-GSH). OTA methyl ester was the major metabolite found in both Caco-2 and HepG2 cells after all treatments. Our results showed that OTA can cause cell damage through several mechanisms and that the OTA exposure time is more important that the dosage in in vitro studies. OTA methyl ester is proposed as an OTA exposure biomarker, although future studies should be conducted.
Collapse
Affiliation(s)
- Cyndia A González-Arias
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain; Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura S/N, Tepic, Nayarit C.P. 63155, Mexico
| | - Sonia Marín
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain
| | - Aurora E Rojas-García
- Laboratorio de Contaminación y Toxicología Ambiental, Secretaría de Investigación y Posgrado, Universidad Autónoma de Nayarit, Ciudad de la Cultura S/N, Tepic, Nayarit C.P. 63155, Mexico
| | - Vicente Sanchis
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain
| | - Antonio J Ramos
- Food Technology Department, Lleida University, UTPV-XaRTA, Agrotecnio Center, Av. Rovira Roure 191, 25198 Lleida, Spain.
| |
Collapse
|
28
|
Sadhukha T, Layek B, Prabha S. Incorporation of lipolysis in monolayer permeability studies of lipid-based oral drug delivery systems. Drug Deliv Transl Res 2017; 8:375-386. [DOI: 10.1007/s13346-017-0383-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Lee JB, Zgair A, Taha DA, Zang X, Kagan L, Kim TH, Kim MG, Yun HY, Fischer PM, Gershkovich P. Quantitative analysis of lab-to-lab variability in Caco-2 permeability assays. Eur J Pharm Biopharm 2017; 114:38-42. [PMID: 28088633 DOI: 10.1016/j.ejpb.2016.12.027] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/28/2016] [Accepted: 12/14/2016] [Indexed: 10/20/2022]
Abstract
In this study, Caco-2 permeability results from different laboratories were compared. Six different sets of apparent permeability coefficient (Papp) values reported in the literature were compared to experimental Papp obtained in our laboratory. The differences were assessed by determining the root mean square error (RMSE) values between the datasets, which reached levels as high as 0.581 for the training set compounds, i.e. ten compounds with known effective human permeability (Peff). The consequences of these differences in Papp for prediction of oral drug absorption were demonstrated by introducing the Papp into the absorption and pharmacokinetics simulation software application GastroPlus™ for prediction of the fraction absorbed (Fa) in humans using calibrated "user-defined permeability models". The RMSE were calculated to assess the differences between the simulated Fa and experimental values reported in the literature. The RMSE for Fa simulated with the permeability model calibrated using experimental Papp from our laboratory was 0.128. When the calibration was performed using Papp from literature datasets, the RMSE values for Fa were higher in all cases except one. This study shows quantitative lab-to-lab variability of Caco-2 permeability results and the potential consequences this can have in the use of these results for predicting intestinal absorption of drugs.
Collapse
Affiliation(s)
- Jong Bong Lee
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Atheer Zgair
- School of Pharmacy, University of Nottingham, Nottingham, UK; College of Pharmacy, University of Anbar, Anbar, Iraq
| | - Dhiaa A Taha
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Xiaowei Zang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Leonid Kagan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Tae Hwan Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Min Gi Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Peter M Fischer
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | |
Collapse
|
30
|
Yamada M, Takei M, Suzuki E, Takakusa H, Kotsuma M, Washio T, Murayama N, Inoue SI, Izumi T. Pharmacokinetics, distribution, and disposition of esaxerenone, a novel, highly potent and selective non-steroidal mineralocorticoid receptor antagonist, in rats and monkeys. Xenobiotica 2016; 47:1090-1103. [PMID: 27866463 DOI: 10.1080/00498254.2016.1263766] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
1. Esaxerenone (CS-3150) is a novel non-steroidal mineralocorticoid receptor antagonist. The pharmacokinetics, tissue distribution, excretion, and metabolism of esaxerenone were evaluated in rats and monkeys. 2. Following intravenous dosing of esaxerenone at 0.1-3 mg/kg, the total body clearance and the volume of distribution were 3.53-6.69 mL/min/kg and 1.47-2.49 L/kg, respectively, in rats, and 2.79-3.69 mL/min/kg and 1.34-1.54 L/kg, respectively, in monkeys. The absolute oral bioavailability was 61.0-127% in rats and 63.7-73.8% in monkeys. 3. After oral administration of [14C]esaxerenone, the radioactivity was distributed widely to tissues, with the exception of a low distribution to the central nervous system. Both in rats and in monkeys, following oral administration of [14C]esaxerenone the main excretion route of the radioactivity was feces. 4. Five initial metabolic pathways in rats and monkeys were proposed to be N-dealkylation, carboxylation, hydroxymethylation, O-glucuronidation, and O-sulfation. The oxidized metabolism was predominant in rats, while both oxidation and glucuronidation were predominant in monkeys.
Collapse
Affiliation(s)
- Makiko Yamada
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Makoto Takei
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Eiko Suzuki
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Hideo Takakusa
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Masakatsu Kotsuma
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Takuo Washio
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Nobuyuki Murayama
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Shin-Ichi Inoue
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| | - Takashi Izumi
- a Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd. , Tokyo , Japan
| |
Collapse
|
31
|
Bale SS, Moore L, Yarmush M, Jindal R. Emerging In Vitro Liver Technologies for Drug Metabolism and Inter-Organ Interactions. TISSUE ENGINEERING PART B-REVIEWS 2016; 22:383-394. [PMID: 27049038 DOI: 10.1089/ten.teb.2016.0031] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In vitro liver models provide essential information for evaluating drug metabolism, metabolite formation, and hepatotoxicity. Interfacing liver models with other organ models could provide insights into the desirable as well as unintended systemic side effects of therapeutic agents and their metabolites. Such information is invaluable for drug screening processes particularly in the context of secondary organ toxicity. While interfacing of liver models with other organ models has been achieved, platforms that effectively provide human-relevant precise information are needed. In this concise review, we discuss the current state-of-the-art of liver-based multiorgan cell culture platforms primarily from a drug and metabolite perspective, and highlight the importance of media-to-cell ratio in interfacing liver models with other organ models. In addition, we briefly discuss issues related to development of optimal liver models that include recent advances in hepatic cell lines, stem cells, and challenges associated with primary hepatocyte-based liver models. Liver-based multiorgan models that achieve physiologically relevant coupling of different organ models can have a broad impact in evaluating drug efficacy and toxicity, as well as mechanistic investigation of human-relevant disease conditions.
Collapse
Affiliation(s)
- Shyam Sundhar Bale
- 1 Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children , Boston, Massachusetts
| | - Laura Moore
- 1 Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children , Boston, Massachusetts
| | - Martin Yarmush
- 1 Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children , Boston, Massachusetts.,2 Department of Biomedical Engineering, Rutgers University , Piscataway, New Jersey
| | - Rohit Jindal
- 1 Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children , Boston, Massachusetts
| |
Collapse
|
32
|
|
33
|
Shen C, Meng Q, Zhang G. Design of 3D printed insert for hanging culture of Caco-2 cells. Biofabrication 2014; 7:015003. [DOI: 10.1088/1758-5090/7/1/015003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Fan CW, Chen B, Franco I, Lu J, Shi H, Wei S, Wang C, Wu X, Tang W, Roth MG, Williams NS, Hirsch E, Chen C, Lum L. The Hedgehog pathway effector smoothened exhibits signaling competency in the absence of ciliary accumulation. ACTA ACUST UNITED AC 2014; 21:1680-9. [PMID: 25484239 DOI: 10.1016/j.chembiol.2014.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 10/22/2014] [Accepted: 10/30/2014] [Indexed: 01/03/2023]
Abstract
Misactivation of the seven-transmembrane protein Smoothened (Smo) is frequently associated with basal cell carcinoma and medulloblastoma. Cellular exposure to secreted Hedgehog (Hh) protein or oncogenic mutations in Hh pathway components induces Smo accumulation in the primary cilium, an antenna-like organelle with mostly unknown cellular functions. Despite the data supporting an indispensable role of the primary cilium in Smo activation, the mechanistic underpinnings of this dependency remain unclear. Using a cell-membrane-impermeable Smo antagonist (IHR-1), we demonstrate that Smo supplied with a synthetic agonist or activated with oncogenic mutations can signal without ciliary accumulation. Similarly, cells with compromised ciliary Smo trafficking due to loss of the phosphatidylinositol-4-phosphate 3-kinase (PI3K)-C2α retain transcriptional response to an exogenously supplied Smo agonist. These observations suggest that assembly of a Smo-signaling complex in the primary cilium is not a prerequisite for Hh pathway activation driven by Smo agonists or oncogenic Smo molecules.
Collapse
Affiliation(s)
- Chih-Wei Fan
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Baozhi Chen
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Irene Franco
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Jianming Lu
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Heping Shi
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Shuguang Wei
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Changguang Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Xiaofeng Wu
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Wei Tang
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Michael G Roth
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Emilio Hirsch
- Molecular Biotechnology Center, Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - Chuo Chen
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Lawrence Lum
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
35
|
Doak B, Over B, Giordanetto F, Kihlberg J. Oral Druggable Space beyond the Rule of 5: Insights from Drugs and Clinical Candidates. ACTA ACUST UNITED AC 2014; 21:1115-42. [DOI: 10.1016/j.chembiol.2014.08.013] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Larregieu CA, Benet LZ. Distinguishing between the permeability relationships with absorption and metabolism to improve BCS and BDDCS predictions in early drug discovery. Mol Pharm 2014; 11:1335-44. [PMID: 24628254 PMCID: PMC3983369 DOI: 10.1021/mp4007858] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
![]()
The biopharmaceutics classification
system (BCS) and biopharmaceutics
drug distribution classification system (BDDCS) are complementary
classification systems that can improve, simplify, and accelerate
drug discovery, development, and regulatory processes. Drug permeability
has been widely accepted as a screening tool for determining intestinal
absorption via the BCS during the drug development and regulatory
approval processes. Currently, predicting clinically significant drug
interactions during drug development is a known challenge for industry
and regulatory agencies. The BDDCS, a modification of BCS that utilizes
drug metabolism instead of intestinal permeability, predicts drug
disposition and potential drug–drug interactions in the intestine,
the liver, and most recently the brain. Although correlations between
BCS and BDDCS have been observed with drug permeability rates, discrepancies
have been noted in drug classifications between the two systems utilizing
different permeability models, which are accepted as surrogate models
for demonstrating human intestinal permeability by the FDA. Here,
we recommend the most applicable permeability models for improving
the prediction of BCS and BDDCS classifications. We demonstrate that
the passive transcellular permeability rate, characterized by means
of permeability models that are deficient in transporter expression
and paracellular junctions (e.g., PAMPA and Caco-2), will most accurately
predict BDDCS metabolism. These systems will inaccurately predict
BCS classifications for drugs that particularly are substrates of
highly expressed intestinal transporters. Moreover, in this latter
case, a system more representative of complete human intestinal permeability
is needed to accurately predict BCS absorption.
Collapse
Affiliation(s)
- Caroline A Larregieu
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco , San Francisco, California 94143-0912, United States
| | | |
Collapse
|
37
|
Larregieu CA, Benet LZ. Drug discovery and regulatory considerations for improving in silico and in vitro predictions that use Caco-2 as a surrogate for human intestinal permeability measurements. AAPS JOURNAL 2013; 15:483-97. [PMID: 23344793 DOI: 10.1208/s12248-013-9456-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 01/10/2013] [Indexed: 11/30/2022]
Abstract
There is a growing need for highly accurate in silico and in vitro predictive models to facilitate drug discovery and development. Results from in vitro permeation studies across the Caco-2 cell monolayer are commonly used for drug permeability screening in industry and are also accepted as a surrogate for human intestinal permeability measurements by the US FDA to support new drug applications. Countless studies carried out in this cell line with published permeability measurements have enabled the development of many in silico prediction models. We identify several common cases that illustrate how using Caco-2 permeability measurements in these in silico and in vitro predictive models will not correlate with human intestinal permeability and will further lead to inaccuracies in these models. We provide guidelines and recommendations for improving these models to more accurately predict clinically relevant information, thereby enhancing the drug discovery, development, and regulatory approval processes.
Collapse
Affiliation(s)
- Caroline A Larregieu
- Department of Bioengineering & Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, 533 Parnassus Avenue, Room U-68, San Francisco, CA 94143-0912, USA
| | | |
Collapse
|
38
|
Hakkarainen JJ, Pajander J, Laitinen R, Suhonen M, Forsberg MM. Similar molecular descriptors determine the in vitro drug permeability in endothelial and epithelial cells. Int J Pharm 2012; 436:426-43. [DOI: 10.1016/j.ijpharm.2012.06.049] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 06/21/2012] [Accepted: 06/22/2012] [Indexed: 11/26/2022]
|
39
|
Hallifax D, Turlizzi E, Zanelli U, Houston JB. Clearance-dependent underprediction of in vivo intrinsic clearance from human hepatocytes: Comparison with permeabilities from artificial membrane (PAMPA) assay, in silico and caco-2 assay, for 65 drugs. Eur J Pharm Sci 2012; 45:570-4. [DOI: 10.1016/j.ejps.2011.12.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2011] [Revised: 11/14/2011] [Accepted: 12/18/2011] [Indexed: 11/27/2022]
|
40
|
Abstract
In silico tools specifically developed for prediction of pharmacokinetic parameters are of particular interest to pharmaceutical industry because of the high potential of discarding inappropriate molecules during an early stage of drug development itself with consequent saving of vital resources and valuable time. The ultimate goal of the in silico models of absorption, distribution, metabolism, and excretion (ADME) properties is the accurate prediction of the in vivo pharmacokinetics of a potential drug molecule in man, whilst it exists only as a virtual structure. Various types of in silico models developed for successful prediction of the ADME parameters like oral absorption, bioavailability, plasma protein binding, tissue distribution, clearance, half-life, etc. have been briefly described in this chapter.
Collapse
Affiliation(s)
- A K Madan
- Pt. BD Sharma University of Health Sciences, Rohtak, India.
| | | |
Collapse
|
41
|
Chao P, Uss AS, Cheng KC. Use of intrinsic clearance for prediction of human hepatic clearance. Expert Opin Drug Metab Toxicol 2010; 6:189-98. [PMID: 20073997 DOI: 10.1517/17425250903405622] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
IMPORTANCE OF THE FIELD The use of intrinsic metabolic stability/clearance and other in vitro pharmacokinetic data for the selection of drug candidates for clinical evaluation during discovery lead optimization has become one of the primary focuses of research organizations involved in new drug discovery. Using intrinsic clearance determined from human liver microsomal preparations and/or hepatocyte to predict human clearance has become more acceptable. AREAS COVERED IN THIS REVIEW This review focuses on the current methods for determining intrinsic clearance and scaling to predict human hepatic clearance, and novel physiologically-based models for improvement of human hepatic clearance prediction. Published microsomal metabolic stability data and in-house hepatocyte clearance data were compared with published in vivo human hepatic clearance data. Various scaling models and the effect of protein binding were examined. WHAT THE READER WILL GAIN Use of a novel microfluidic model and other physiologically-based models are presented. Microsomal metabolic clearance requires correction for protein binding and in vitro microsomal binding in order to better predict in vivo hepatic clearance of compounds that are mainly eliminated by hepatic metabolism. TAKE HOME MESSAGE Metabolic clearance obtained using hepatocytes may work well in combination with the well-stirred model. Novel models incorporating flow and protein binding in the system may be the most complete models for prediction of human in vivo metabolism.
Collapse
Affiliation(s)
- Piyun Chao
- Hurel Corporation, Bervely Hills, CA 91201, USA
| | | | | |
Collapse
|
42
|
Novik E, Maguire TJ, Chao P, Cheng KC, Yarmush ML. A microfluidic hepatic coculture platform for cell-based drug metabolism studies. Biochem Pharmacol 2009; 79:1036-44. [PMID: 19925779 DOI: 10.1016/j.bcp.2009.11.010] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 11/02/2009] [Accepted: 11/10/2009] [Indexed: 11/29/2022]
Abstract
Within the global pharmaceutical and biotech industries, there is significant interest in identifying in vitro screening systems that are more human-relevant-i.e., that offer greater utility in predicting subcellular and cellular physiological responses in humans in vivo-and that thereby allow investigators to reduce the incidence of costly late-stage failures during pharmaceutical clinical trials, as well as to reduce the use of animals in drug testing. Currently incumbent in vitro screening methods, such as culturing human hepatocytes in suspension, while useful, are limited by a lack of long term cellular function. In order to address this limitation, we have established an integrated, microfluidic, in vitro platform that combines the patented HmuREL((R)) microdevice with a hepatic coculture system. In the present report, we use this platform to study clearance and metabolite generation of a battery of molecular entities. The results show that the flow-based coculture system is capable of clearing, with improved resolution and predictive value, compounds with high, medium, and low clearance values. In addition, when coculture is coupled with flow, higher metabolite production rates are obtained than in static systems.
Collapse
Affiliation(s)
- Eric Novik
- Hurel Corporation, Beverley Hills, California, CA, USA
| | | | | | | | | |
Collapse
|
43
|
Chao P, Maguire T, Novik E, Cheng KC, Yarmush ML. Evaluation of a microfluidic based cell culture platform with primary human hepatocytes for the prediction of hepatic clearance in human. Biochem Pharmacol 2009; 78:625-32. [PMID: 19463793 DOI: 10.1016/j.bcp.2009.05.013] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 05/12/2009] [Accepted: 05/12/2009] [Indexed: 11/30/2022]
Abstract
Integral to the discovery of new pharmaceutical entities is the ability to predict in vivo pharmacokinetic parameters from early stage in vitro data generated prior to the onset of clinical testing. Within the pharmaceutical industry, a whole host of assay methods and mathematical models exist to predict the in vivo pharmacokinetic parameters of drug candidates. One of the most important pharmacokinetic properties of new drug candidates predicted from these methods and models is the hepatic clearance. Current methods, while useful, are still limited in their predictive efficacy. In order to address this issue, we have established a novel microfluidic in vitro culture system, the patented HmuREL device. The device comprises multiple compartments that are designed to be proportional to the physiological architectures and enhanced with the consideration of flow. Here we demonstrate the functionality of the liver-relevant chamber in the HmuREL device, and the feasibility of utilizing our system for predicting hepatic clearance. Cryopreserved human hepatocytes from a single donor were seeded within the HmuREL device to predict the in vivo hepatic clearance (CL(H)) of six marketed model compounds (carbamazepine, caffeine, timolol, sildenafil, imipramine, and buspirone). The intrinsic clearance rates from static culture controls, as well as clearance rates from the HmuREL device were subsequently compared to in vivo data available from the literature.
Collapse
Affiliation(s)
- P Chao
- Hurel Corporation, Beverley Hills, CA, USA
| | | | | | | | | |
Collapse
|
44
|
Yuan Z, Xu H, Wang K, Zhao Z, Hu M. Determination of osthol and its metabolites in a phase I reaction system and the Caco-2 cell model by HPLC-UV and LC-MS/MS. J Pharm Biomed Anal 2008; 49:1226-32. [PMID: 19304430 DOI: 10.1016/j.jpba.2008.12.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 12/01/2008] [Accepted: 12/01/2008] [Indexed: 11/16/2022]
Abstract
A straightforward and sensitive reversed-phase high-performance liquid chromatography (HPLC) assay was developed and validated for the analysis of osthol and its phase I metabolites (internal standard: umbelliferone). The method was validated for the determination of osthol with respect to selectivity, precision, linearity, limit of detection, recovery, and stability. The linear response range was 0.47-60 microM, and the average recoveries ranged from 98 to 101%. The inter-day and intra-day relative standard deviations were both less than 5%. Using this method, we showed that more than 80% of osthol was metabolized in 20 min in a phase I metabolic reaction system. Transport experiments in the Caco-2 cell culture model indicated that osthol was easily absorbed with high absorptive permeability (>10 x 10(-6)cm/s). The permeability did not display concentration-dependence or vectorial-dependence and is mildly temperature sensitive (activation energy less than 10 kcal/mol), indicating passive mechanism of transport. When analyzed by LC-MS/MS, five metabolites were detected in a phase I reaction system and in the receiver side of a modified Caco-2 cell model, which was supplemented with the phase I reaction system. The major metabolites appeared to be desmethyl-osthol and multiple isomers of dehydro-osthol. In conclusion, a likely cause of poor osthol bioavailability is rapid phase I metabolism via the cytochrome P450 pathways.
Collapse
Affiliation(s)
- Zhenting Yuan
- No. 230 Hospital in Dandong, Dandong 118000, PR China
| | | | | | | | | |
Collapse
|