1
|
Bacon J, Kitchel H, Stutz J, Chen JH, Smith A, Van Horn RD, Moreland C, Abraham T, Baker T, Aihara E, Hillgren K. Porcine intestinal organoids cultured in an organ-on-a-chip microphysiological system. Biochem Biophys Rep 2025; 42:102036. [PMID: 40421277 PMCID: PMC12104630 DOI: 10.1016/j.bbrep.2025.102036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/28/2025] Open
Abstract
Preclinical studies are a vital component of pharmaceutical development and improvements in the predictive value of in vitro studies are essential. Organ-on-a-chip in vitro models are a recent advancement in the pursuit of improved reproduction of in vivo tissue complexity. Here, we report the development and characterization of porcine intestinal cells from organoids on chips with microfluid dynamics and peristaltic-like strain in a microphysiological system. Intestinal epithelial cells were grown on a porous membrane as a co-culture with human intestinal microvascular endothelial cells for up to 12 days. These cultures formed villi-like structures and established a tight barrier replete with F-actin and tight junctions. A demarcated region of the epithelial cells was in an actively proliferative stage, reminiscent of intestinal crypts. The intestinal epithelial cell growth was characterized for the presence of enterocytes, goblet cells and enteroendocrine cells. Notable drug transporters and CYP450 metabolic activity were present in these cultures. The organoid chip maintained barrier function as the paracellular permeability was low. In contrast, the permeability enhancer, sodium caprate (C10), increased the apparent permeability of molecular weight marker compounds by 2- to 3-fold, and upon removal of C10, the barrier was shown to be recovered. The porcine intestinal chip represents a new in vitro model with potential application in multiple aspects of pharmaceutical testing including drug metabolism, drug transporters and safety.
Collapse
Affiliation(s)
- James Bacon
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Halie Kitchel
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - John Stutz
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Jack Hua Chen
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Aaron Smith
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Robert D. Van Horn
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | | | - Trent Abraham
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | | | - Eitaro Aihara
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Kathleen Hillgren
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| |
Collapse
|
2
|
Imaoka T, Onuki-Nagasaki R, Kimura H, Tai K, Ishii M, Nozue A, Kaisaki I, Hoshi M, Watanabe K, Maeda K, Kamizono T, Yoshioka T, Fujimoto T, Satoh T, Nakamura H, Ando O, Kusuhara H, Ito Y. Development of a novel gut microphysiological system that facilitates assessment of drug absorption kinetics in gut. Sci Rep 2024; 14:29921. [PMID: 39622870 PMCID: PMC11612460 DOI: 10.1038/s41598-024-80946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
There is an urgent need for novel methods that can accurately predict intestinal absorption of orally administered drugs in humans. This study aimed to evaluate the potential of a novel gut microphysiological system (MPS), gut MPS/Fluid3D-X, to assess the intestinal absorption of drugs in humans. The gut MPS/Fluid3D-X model was constructed using a newly developed flow-controllable and dimethylpolysiloxane-free MPS device (Fluid3D-X®). Human induced pluripotent stem cells-derived small intestinal epithelial cells were employed in this model, which exhibited key characteristics of the human absorptive epithelial cells of the small intestine, including the expression of key gene transcripts responsible for drug transport and metabolism, and the presence of dome-like protrusions in the primary intestinal epithelium under air-liquid interface culture conditions. Functional studies of transporters in the constructed model demonstrated basal-to-apical directional transport of sulfasalazine and quinidine, substrates of the active efflux transporters breast cancer resistance protein and P-glycoprotein, respectively, which were diminished by inhibitors. Furthermore, a cytochrome P450 (CYP) 3A inhibitor increased the apical-to-basal transport of midazolam, a typical CYP3A4 substrate, and reduced metabolite formation. These results suggest that gut MPS/Fluid3D-X has the potential to assess the intestinal absorption of small-molecule drugs.
Collapse
Grants
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
- JP17be0304204, JP17be0304101, P22be1004101 and JP22be1004301 The Japanese Agency for Medical Research and Development
Collapse
Affiliation(s)
- Tomoki Imaoka
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Reiko Onuki-Nagasaki
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Hiroshi Kimura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, 259-1292, Kanagawa, Japan
| | - Kempei Tai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo- ku, Tokyo, 113-0033, Japan
| | - Mitsuharu Ishii
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Ayaka Nozue
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Ikuko Kaisaki
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo- ku, Tokyo, 113-0033, Japan
| | - Misa Hoshi
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kengo Watanabe
- Drug Metabolism & Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd, 1-2-58, Hiromachi, Shinagawa-ku, Tokyo, 140-8710, Japan
| | - Kazuya Maeda
- Kitasato University School of Pharmacy, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Takashi Kamizono
- Tokyo Ohka Kogyo Co. Ltd, Samukawa-machi, Koza-gun, Tabata, 1590, 253-0114, Kanagawa, Japan
| | - Takahiro Yoshioka
- Tokyo Ohka Kogyo Co. Ltd, Samukawa-machi, Koza-gun, Tabata, 1590, 253-0114, Kanagawa, Japan
| | - Takashi Fujimoto
- Tokyo Ohka Kogyo Co. Ltd, Samukawa-machi, Koza-gun, Tabata, 1590, 253-0114, Kanagawa, Japan
| | - Taku Satoh
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Hiroko Nakamura
- Micro/Nano Technology Center, Tokai University, 4-1-1 Kitakaname, Hiratsuka, 259-1292, Kanagawa, Japan
| | - Osamu Ando
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Hiroyuki Kusuhara
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo- ku, Tokyo, 113-0033, Japan
| | - Yuzuru Ito
- Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan.
| |
Collapse
|
3
|
Volpe DA. Application of transporter assays for drug discovery and development: an update of the literature. Expert Opin Drug Discov 2024; 19:1247-1257. [PMID: 39105537 DOI: 10.1080/17460441.2024.2387790] [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: 05/20/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
INTRODUCTION Determining whether a new drug is a substrate, inhibitor or inducer of efflux or uptake membrane transporters has become a routine process during drug discovery and development. In vitro assays are utilized to establish whether a new drug has the potential to be an object (substrate) or precipitant (inhibitor, inducer) in transporter-mediated clinical drug-drug interactions. The findings from these in vitro experiments are then used to determine whether further in vivo drug interaction studies are necessary for a new drug. AREAS COVERED This article provides an update on in vitro transporter assays, focusing on new uses of transfected cells, time-dependent inhibition, transporter induction, and complex model systems. EXPERT OPINION The newer in vitro assays add to the toolbox in defining new drugs as transporter substrates, inhibitors, or inducers. Complex models such as spheroids, organoids, and microphysiological systems require standardization and further research with model transporter substrates and inhibitors. In drug discovery, the more traditional transporter assays may be employed as substrate and inhibitor screening assays. In drug development, more complex cell models can be employed in later drug development to better understand how transporter(s) are involved in the absorption, distribution, and excretion of new drugs.
Collapse
Affiliation(s)
- Donna A Volpe
- Division of Applied Regulatory Science, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
4
|
Singla P, Broughton T, Sullivan MV, Garg S, Berlinguer‐Palmini R, Gupta P, Smith KJ, Gardner B, Canfarotta F, Turner NW, Velliou E, Amarnath S, Peeters M. Double Imprinted Nanoparticles for Sequential Membrane-to-Nuclear Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309976. [PMID: 38973256 PMCID: PMC11423068 DOI: 10.1002/advs.202309976] [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: 02/15/2024] [Revised: 06/14/2024] [Indexed: 07/09/2024]
Abstract
Efficient and site-specific delivery of therapeutics drugs remains a critical challenge in cancer treatment. Traditional drug nanocarriers such as antibody-drug conjugates are not generally accessible due to their high cost and can lead to serious side effects including life-threatening allergic reactions. Here, these problems are overcome via the engineering of supramolecular agents that are manufactured with an innovative double imprinting approach. The developed molecularly imprinted nanoparticles (nanoMIPs) are targeted toward a linear epitope of estrogen receptor alfa (ERα) and loaded with the chemotherapeutic drug doxorubicin. These nanoMIPs are cost-effective and rival the affinity of commercial antibodies for ERα. Upon specific binding of the materials to ERα, which is overexpressed in most breast cancers (BCs), nuclear drug delivery is achieved via receptor-mediated endocytosis. Consequentially, significantly enhanced cytotoxicity is elicited in BC cell lines overexpressing ERα, paving the way for precision treatment of BC. Proof-of-concept for the clinical use of the nanoMIPs is provided by evaluating their drug efficacy in sophisticated three-dimensional (3D) cancer models, which capture the complexity of the tumor microenvironment in vivo without requiring animal models. Thus, these findings highlight the potential of nanoMIPs as a promising class of novel drug compounds for use in cancer treatment.
Collapse
Affiliation(s)
- Pankaj Singla
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Thomas Broughton
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- NIHR, Biomedical Research CentreNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Mark V. Sullivan
- Department of ChemistryUniversity of SheffieldDainton BuildingSheffieldS3 7HFUK
| | - Saweta Garg
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Rolando Berlinguer‐Palmini
- The Bio‐Imaging Unit, Medical SchoolNewcastle UniversityWilliam Leech BuildingNewcastle Upon TyneNE2 4HHUK
| | - Priyanka Gupta
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional ScienceUniversity College LondonLondonW1W 7TYUK
| | - Katie J Smith
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Ben Gardner
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | | | - Nicholas W. Turner
- Department of ChemistryUniversity of SheffieldDainton BuildingSheffieldS3 7HFUK
| | - Eirini Velliou
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional ScienceUniversity College LondonLondonW1W 7TYUK
| | - Shoba Amarnath
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- Immune Regulation Laboratory, NU Biosciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
- NIHR, Biomedical Research CentreNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| | - Marloes Peeters
- Department of Chemical EngineeringThe University of ManchesterEngineering building A, East Booth Street, Oxford RoadManchesterM13 9PLUK
- School of EngineeringNewcastle UniversityMerz Court, Claremont RoadNewcastle Upon TyneNE1 7RUUK
- Center for Cancer Research, NU Cancer, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneNE2 4HHUK
| |
Collapse
|
5
|
Modi PS, Singh A, Chaturvedi A, Agarwal S, Dutta R, Nayak R, Singh AK. Tissue chips as headway model and incitement technology. Synth Syst Biotechnol 2024; 10:86-101. [PMID: 39286054 PMCID: PMC11403008 DOI: 10.1016/j.synbio.2024.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Tissue on a chip or organ-on-chip (OOC) is a technology that's dignified to form a transformation in drug discovery through the use of advanced platforms. These are 3D in-vitro cell culture models that mimic micro-environment of human organs or tissues on artificial microstructures built on a portable microfluidic chip without involving sacrificial humans or animals. This review article aims to offer readers a thorough and insightful understanding of technology. It begins with an in-depth understanding of chip design and instrumentation, underlining its pivotal role and the imperative need for its development in the modern scientific landscape. The review article explores into the myriad applications of OOC technology, showcasing its transformative impact on fields such as radiobiology, drug discovery and screening, and its pioneering use in space research. In addition to highlighting these diverse applications, the article provides a critical analysis of the current challenges that OOC technology faces. It examines both the biological and technical limitations that hinder its progress and efficacy and discusses the potential advancements and innovations that could drive the OOC technology forward. Through this comprehensive review, readers will gain a deep appreciation of the significance, capabilities, and evolving landscape of OOC technology.
Collapse
Affiliation(s)
- Prerna Suchitan Modi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Abhishek Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Awyang Chaturvedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Shailly Agarwal
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Raghav Dutta
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| | - Alok Kumar Singh
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, Uttar Pradesh, India
| |
Collapse
|
6
|
Tomlinson L, Ramsden D, Leite SB, Beken S, Bonzo JA, Brown P, Candarlioglu PL, Chan TS, Chen E, Choi CK, David R, Delrue N, Devine PJ, Ford K, Garcia MI, Gosset JR, Hewitt P, Homan K, Irrechukwu O, Kopec AK, Liras JL, Mandlekar S, Raczynski A, Sadrieh N, Sakatis MZ, Siegel J, Sung K, Sunyovszki I, Van Vleet TR, Ekert JE, Hardwick RN. Considerations from an International Regulatory and Pharmaceutical Industry (IQ MPS Affiliate) Workshop on the Standardization of Complex In Vitro Models in Drug Development. Adv Biol (Weinh) 2024; 8:e2300131. [PMID: 37814378 DOI: 10.1002/adbi.202300131] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/08/2023] [Indexed: 10/11/2023]
Abstract
In May 2022, there is an International Regulatory and Pharmaceutical Industry (Innovation and Quality [IQ] Microphysiological Systems [MPS] Affiliate) Workshop on the standardization of complex in vitro models (CIVMs) in drug development. This manuscript summarizes the discussions and conclusions of this joint workshop organized and executed by the IQ MPS Affiliate and the United States Food and Drug Administration (FDA). A key objective of the workshop is to facilitate discussions around opportunities and/or needs for standardization of MPS and chart potential pathways to increase model utilization in the context of regulatory decision making. Participation in the workshop included 200 attendees from the FDA, IQ MPS Affiliate, and 26 global regulatory organizations and affiliated parties representing Europe, Japan, and Canada. It is agreed that understanding global perspectives regarding the readiness of CIVM/MPS models for regulatory decision making and potential pathways to gaining acceptance is useful to align on globally. The obstacles are currently too great to develop standards for every context of use (COU). Instead, it is suggested that a more tractable approach may be to think of broadly applicable standards that can be applied regardless of COU and/or organ system. Considerations and next steps for this effort are described.
Collapse
Affiliation(s)
| | | | | | - Sonja Beken
- Federal Agency for Medicines and Health Products, Brussels, 1210, Belgium
| | - Jessica A Bonzo
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Paul Brown
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | | | - Tom S Chan
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, 06877, USA
| | - Eugene Chen
- DMPK, Genentech, South San Francisco, CA, 94080, USA
| | - Colin K Choi
- Preclinical Safety, Biogen, Cambridge, MA, 02142, USA
| | - Rhiannon David
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Nathalie Delrue
- Organisation for Economic Co-operation and Development, Paris, 75016, France
| | - Patrick J Devine
- Discovery Toxicology, Bristol Myers Squibb, San Diego, CA, 09130, USA
| | - Kevin Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Martha Iveth Garcia
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Kimberly Homan
- Complex in Vitro Systems Group, Genentech, South San Francisco, CA, 94080, USA
| | - Onyi Irrechukwu
- Preclinical Sciences and Translational Safety, Johnson and Johnson Innovation Medicine, Spring House, PA, 19002, USA
| | - Anna K Kopec
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, 06340, USA
| | - Jennifer L Liras
- Pharmacokinetics, Dynamics & Metabolism-Medicine Design, Pfizer, Cambridge, MA, 02139, USA
| | - Sandhya Mandlekar
- Clinical Pharmacology, Genentech, South San Francisco, CA, 94080, USA
| | - Arek Raczynski
- Preclinical Safety Assessment, Vertex Pharmaceuticals Inc., Boston, MA, 02210, USA
| | - Nakissa Sadrieh
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Melanie Z Sakatis
- Non-Clinical Safety, In Vitro/In Vivo Translation, GSK R&D, Ware, SG12 9TJ, UK
| | - Jeffrey Siegel
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Kyung Sung
- Center for Biologics Evaluation and Research, Office of Cellular Therapy and Human Tissue, Cellular and Tissue Therapy Branch, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ilona Sunyovszki
- Translational Cellular Sciences, Biogen, Cambridge, MA, 02142, USA
| | | | | | | |
Collapse
|
7
|
Carius P, Weinelt FA, Cantow C, Holstein M, Teitelbaum AM, Cui Y. Addressing the ADME Challenges of Compound Loss in a PDMS-Based Gut-on-Chip Microphysiological System. Pharmaceutics 2024; 16:296. [PMID: 38543190 PMCID: PMC10974294 DOI: 10.3390/pharmaceutics16030296] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 12/13/2024] Open
Abstract
Microphysiological systems (MPSs) are promising in vitro technologies for physiologically relevant predictions of the human absorption, distribution, metabolism, and excretion (ADME) properties of drug candidates. However, polydimethylsiloxane (PDMS), a common material used in MPSs, can both adsorb and absorb small molecules, thereby compromising experimental results. This study aimed to evaluate the feasibility of using the PDMS-based Emulate gut-on-chip to determine the first-pass intestinal drug clearance. In cell-free PDMS organ-chips, we assessed the loss of 17 drugs, among which testosterone was selected as a model compound for further study based on its substantial ad- and absorptions to organ chips and its extensive first-pass intestinal metabolism with well-characterized metabolites. A gut-on-chip model consisting of epithelial Caco-2 cells and primary human umbilical vein endothelial cells (HUVECs) was established. The barrier integrity of the model was tested with reference compounds and inhibition of drug efflux. Concentration-time profiles of testosterone were measured in cell-free organ chips and in gut-on-chip models. A method to deduce the metabolic clearance was provided. Our results demonstrate that metabolic clearance can be determined with PDMS-based MPSs despite substantial compound loss to the chip. Overall, this study offers a practical protocol to experimentally assess ADME properties in PDMS-based MPSs.
Collapse
Affiliation(s)
| | | | | | | | | | - Yunhai Cui
- Department Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, 88400 Biberach, Germany; (P.C.); (F.A.W.); (C.C.); (M.H.); (A.M.T.)
| |
Collapse
|
8
|
Kalia V, Baccarelli AA, Happel C, Hollander JA, Jukic AM, McAllister KA, Menon R, Merrick BA, Milosavljevic A, Ravichandran LV, Roth ME, Subramanian A, Tyson FL, Worth L, Shaughnessy DT. Seminar: Extracellular Vesicles as Mediators of Environmental Stress in Human Disease. ENVIRONMENTAL HEALTH PERSPECTIVES 2023; 131:104201. [PMID: 37861803 PMCID: PMC10588739 DOI: 10.1289/ehp12980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Extracellular vesicles (EVs), membrane-bound particles containing a variety of RNA types, DNA, proteins, and other macromolecules, are now appreciated as an important means of communication between cells and tissues, both in normal cellular physiology and as a potential indicator of cellular stress, environmental exposures, and early disease pathogenesis. Extracellular signaling through EVs is a growing field of research for understanding fundamental mechanisms of health and disease and for the potential for biomarker discovery and therapy development. EVs are also known to play important roles in mediating the effects of exposure to environmental stress. OBJECTIVES This seminar addresses the application of new tools and approaches for EV research, developed in part through the National Institutes of Health (NIH) Extracellular RNA Communication Program, and reflects presentations and discussions from a workshop held 27-28 September 2021 by the National Institute of Environmental Health Sciences (NIEHS) and the National Center for Advancing Translational Sciences (NCATS) on "Extracellular Vesicles, Exosomes, and Cell-Cell Signaling in Response to Environmental Stress." The panel of experts discussed current research on EVs and environmental exposures, highlighted recent advances in EV isolation and characterization, and considered research gaps and opportunities toward identifying and characterizing the roles for EVs in environmentally related diseases, as well as the current challenges and opportunities in this field. DISCUSSION The authors discuss the application of new experimental models, particularly organ-on-chip (OOC) systems and in vitro approaches and how these have the potential to extend findings in population-based studies of EVs in exposure-related diseases. Given the complex challenges of identifying cell-specific EVs related to environmental exposures, as well as the general heterogeneity and variability in EVs in blood and other accessible biological samples, there is a critical need for rigorous reporting of experimental methods and validation studies. The authors note that these efforts, combined with cross-disciplinary approaches, would ensure that future research efforts in environmental health studies on EV biomarkers are rigorous and reproducible. https://doi.org/10.1289/EHP12980.
Collapse
Affiliation(s)
- Vrinda Kalia
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Christine Happel
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), U.S. Department of Health and Human Services (DHHS), Bethesda, Maryland, USA
| | - Jonathan A. Hollander
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Anne Marie Jukic
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Kimberly A. McAllister
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Basic Science and Translational Research, University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Bruce A. Merrick
- Division of Translational Toxicology, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | | | - Lingamanaidu V. Ravichandran
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Matthew E. Roth
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Anita Subramanian
- Division of Intramural Research, NIEHS, NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Frederick L. Tyson
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Leroy Worth
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| | - Daniel T. Shaughnessy
- Division of Extramural Research and Training, National Institute of Environmental Health Sciences (NIEHS), NIH, DHHS, Research Triangle Park, North Carolina, USA
| |
Collapse
|
9
|
Fu J, Qiu H, Tan CS. Microfluidic Liver-on-a-Chip for Preclinical Drug Discovery. Pharmaceutics 2023; 15:pharmaceutics15041300. [PMID: 37111785 PMCID: PMC10141038 DOI: 10.3390/pharmaceutics15041300] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/31/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Drug discovery is an expensive, long, and complex process, usually with a high degree of uncertainty. In order to improve the efficiency of drug development, effective methods are demanded to screen lead molecules and eliminate toxic compounds in the preclinical pipeline. Drug metabolism is crucial in determining the efficacy and potential side effects, mainly in the liver. Recently, the liver-on-a-chip (LoC) platform based on microfluidic technology has attracted widespread attention. LoC systems can be applied to predict drug metabolism and hepatotoxicity or to investigate PK/PD (pharmacokinetics/pharmacodynamics) performance when combined with other artificial organ-on-chips. This review discusses the liver physiological microenvironment simulated by LoC, especially the cell compositions and roles. We summarize the current methods of constructing LoC and the pharmacological and toxicological application of LoC in preclinical research. In conclusion, we also discussed the limitations of LoC in drug discovery and proposed a direction for improvement, which may provide an agenda for further research.
Collapse
Affiliation(s)
- Jingyu Fu
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| | - Hailong Qiu
- Tianjin Key Laboratory of Functional Crystal Materials, Institute of Functional Crystal, Tianjin University of Technology, Tianjin 300384, China
| | - Cherie S Tan
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin 300072, China
| |
Collapse
|
10
|
Gökçe F, Kaestli A, Lohasz C, de Geus M, Kaltenbach H, Renggli K, Bornhauser B, Hierlemann A, Modena M. Microphysiological Drug-Testing Platform for Identifying Responses to Prodrug Treatment in Primary Leukemia. Adv Healthc Mater 2023; 12:e2202506. [PMID: 36651229 PMCID: PMC11469234 DOI: 10.1002/adhm.202202506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 12/20/2022] [Indexed: 01/19/2023]
Abstract
Despite increasing survival rates of pediatric leukemia patients over the past decades, the outcome of some leukemia subtypes has remained dismal. Drug sensitivity and resistance testing on patient-derived leukemia samples provide important information to tailor treatments for high-risk patients. However, currently used well-based drug screening platforms have limitations in predicting the effects of prodrugs, a class of therapeutics that require metabolic activation to become effective. To address this issue, a microphysiological drug-testing platform is developed that enables co-culturing of patient-derived leukemia cells, human bone marrow mesenchymal stromal cells, and human liver microtissues within the same microfluidic platform. This platform also enables to control the physical interaction between the diverse cell types. Herein, it is made possible to recapitulate hepatic prodrug activation of ifosfamide in their platform, which is very difficult in traditional well-based assays. By testing the susceptibility of primary patient-derived leukemia samples to the prodrug ifosfamide, sample-specific sensitivities to ifosfamide in primary leukemia samples are identified. The microfluidic platform is found to enable the recapitulation of physiologically relevant conditions and the testing of prodrugs including short-lived and unstable metabolites. The platform holds great potential for clinical translation and precision chemotherapy selection.
Collapse
Affiliation(s)
- Furkan Gökçe
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Alicia Kaestli
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Christian Lohasz
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Martina de Geus
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | | | - Kasper Renggli
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Beat Bornhauser
- Children's Research CenterUniversity Children's Hospital ZurichZurichZH, 8008Switzerland
| | - Andreas Hierlemann
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| | - Mario Modena
- Department of Biosystems Science and EngineeringETH ZurichBaselBS, 4058Switzerland
| |
Collapse
|
11
|
Catelli Rocha Torres L, Giovanini de Oliveira Sartori A, Paula de Souza Silva A, Matias de Alencar S. Bioaccessibility and uptake/epithelial transport of vitamin E: discoveries and challenges of in vitro and ex vivo assays. Food Res Int 2022; 162:112143. [DOI: 10.1016/j.foodres.2022.112143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/01/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
|
12
|
Giacomini KM, Yee SW, Koleske ML, Zou L, Matsson P, Chen EC, Kroetz DL, Miller MA, Gozalpour E, Chu X. New and Emerging Research on Solute Carrier and ATP Binding Cassette Transporters in Drug Discovery and Development: Outlook From the International Transporter Consortium. Clin Pharmacol Ther 2022; 112:540-561. [PMID: 35488474 PMCID: PMC9398938 DOI: 10.1002/cpt.2627] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023]
Abstract
Enabled by a plethora of new technologies, research in membrane transporters has exploded in the past decade. The goal of this state-of-the-art article is to describe recent advances in research on membrane transporters that are particularly relevant to drug discovery and development. This review covers advances in basic, translational, and clinical research that has led to an increased understanding of membrane transporters at all levels. At the basic level, we describe the available crystal structures of membrane transporters in both the solute carrier (SLC) and ATP binding cassette superfamilies, which has been enabled by the development of cryogenic electron microscopy methods. Next, we describe new research on lysosomal and mitochondrial transporters as well as recently deorphaned transporters in the SLC superfamily. The translational section includes a summary of proteomic research, which has led to a quantitative understanding of transporter levels in various cell types and tissues and new methods to modulate transporter function, such as allosteric modulators and targeted protein degraders of transporters. The section ends with a review of the effect of the gut microbiome on modulation of transporter function followed by a presentation of 3D cell cultures, which may enable in vivo predictions of transporter function. In the clinical section, we describe new genomic and pharmacogenomic research, highlighting important polymorphisms in transporters that are clinically relevant to many drugs. Finally, we describe new clinical tools, which are becoming increasingly available to enable precision medicine, with the application of tissue-derived small extracellular vesicles and real-world biomarkers.
Collapse
Affiliation(s)
- Kathleen M. Giacomini
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Sook W. Yee
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Megan L. Koleske
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Ling Zou
- Pharmacokinetics and Drug MetabolismAmgen Inc.South San FranciscoCaliforniaUSA
| | - Pär Matsson
- Department of PharmacologySahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Eugene C. Chen
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Deanna L. Kroetz
- Department of Bioengineering and Therapeutic SciencesUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Miles A. Miller
- Center for Systems BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Elnaz Gozalpour
- Drug Safety and MetabolismIMED Biotech UnitSafety and ADME Translational Sciences DepartmentAstraZeneca R&DCambridgeUK
| | - Xiaoyan Chu
- Department of ADME and Discovery ToxicologyMerck & Co. IncKenilworthNew JerseyUSA
| |
Collapse
|
13
|
Mahadeo A, Yeung CK, Himmelfarb J, Kelly EJ. Kidney microphysiological models for nephrotoxicity assessment. CURRENT OPINION IN TOXICOLOGY 2022; 30:100341. [PMID: 35495549 PMCID: PMC9053105 DOI: 10.1016/j.cotox.2022.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Nephrotoxicity testing is an important step in preclinical development of new molecular entities (NMEs) and has traditionally been performed in 2-D cell culture systems and animal models. However, 2-D culture systems fail to replicate complex in vivo microenvironment and animal models face interspecies differences including the overexpression of drug transporters. In the last decade, 3-D microphysiological systems (MPS) have been developed to address these concerns. Here, we review recent advancements in kidney MPS and their application in drug-induced toxicity testing and kidney disease research. We find that current research is making significant progress addressing MPS limitations such as throughput, incorporating various regions of the nephron such as the glomerulus, and successfully modeling and predicting clinically relevant nephrotoxicity of current and new drugs.
Collapse
Affiliation(s)
- Anish Mahadeo
- Department of Pharmaceutics, University of Washington, Seattle, WA
| | - Catherine K Yeung
- Department of Pharmacy, University of Washington, Seattle, WA
- The Kidney Research Institute, University of Washington, Seattle, WA
| | | | - Edward J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, WA
- The Kidney Research Institute, University of Washington, Seattle, WA
| |
Collapse
|
14
|
Dunvald ACD, Järvinen E, Mortensen C, Stage TB. Clinical and Molecular Perspectives on Inflammation-Mediated Regulation of Drug Metabolism and Transport. Clin Pharmacol Ther 2021; 112:277-290. [PMID: 34605009 DOI: 10.1002/cpt.2432] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
Inflammation is a possible cause of variability in drug response and toxicity due to altered regulation in drug-metabolizing enzymes and transporters (DMETs) in humans. Here, we evaluate the clinical and in vitro evidence on inflammation-mediated modulation of DMETs, and the impact on drug metabolism in humans. Furthermore, we identify and discuss the gaps in our current knowledge. A systematic literature search on PubMed, Embase, and grey literature was performed in the period of February to September 2020. A total of 203 papers was included. In vitro studies in primary human hepatocytes revealed strong evidence that CYP3A4 is strongly downregulated by inflammatory cytokines IL-6 and IL-1β. CYP1A2, CYP2C9, CYP2C19, and CYP2D6 were downregulated to a lesser extent. In clinical studies, acute and chronic inflammatory diseases were observed to cause downregulation of CYP enzymes in a similar pattern. However, there is no clear correlation between in vitro studies and clinical studies, mainly because most in vitro studies use supraphysiological cytokine doses. Moreover, clinical studies demonstrate considerable variability in terms of methodology and inconsistencies in evaluation of the inflammatory state. In conclusion, we find inflammation and pro-inflammatory cytokines to be important factors in regulation of drug-metabolizing enzymes and transporters. The observed downregulation is clinically relevant, and we emphasize caution when treating patients in an inflammatory state with narrow therapeutic index drugs. Further research is needed to identify the full extent of inflammation-mediated changes in DMETs and to further support personalized medicine.
Collapse
Affiliation(s)
- Ann-Cathrine Dalgård Dunvald
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense C, Denmark
| | - Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense C, Denmark
| | - Christina Mortensen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense C, Denmark
| | - Tore B Stage
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|