1
|
Chen DY, Turcinovic J, Feng S, Kenney DJ, Chin CV, Choudhary MC, Conway HL, Semaan M, Close BJ, Tavares AH, Seitz S, Khan N, Kapell S, Crossland NA, Li JZ, Douam F, Baker SC, Connor JH, Saeed M. Cell culture systems for isolation of SARS-CoV-2 clinical isolates and generation of recombinant virus. iScience 2023; 26:106634. [PMID: 37095858 PMCID: PMC10083141 DOI: 10.1016/j.isci.2023.106634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 02/22/2023] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
A simple and robust cell culture system is essential for generating authentic SARS-CoV-2 stocks for evaluation of viral pathogenicity, screening of antiviral compounds, and preparation of inactivated vaccines. Evidence suggests that Vero E6, a cell line commonly used in the field to grow SARS-CoV-2, does not support efficient propagation of new viral variants and triggers rapid cell culture adaptation of the virus. We generated a panel of 17 human cell lines overexpressing SARS-CoV-2 entry factors and tested their ability to support viral infection. Two cell lines, Caco-2/AT and HuH-6/AT, demonstrated exceptional susceptibility, yielding highly concentrated virus stocks. Notably, these cell lines were more sensitive than Vero E6 cells in recovering SARS-CoV-2 from clinical specimens. Further, Caco-2/AT cells provided a robust platform for producing genetically reliable recombinant SARS-CoV-2 through a reverse genetics system. These cellular models are a valuable tool for the study of SARS-CoV-2 and its continuously emerging variants.
Collapse
Affiliation(s)
- Da-Yuan Chen
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Jacquelyn Turcinovic
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Shuchen Feng
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Devin J. Kenney
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Chue Vin Chin
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Manish C. Choudhary
- Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Hasahn L. Conway
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Marc Semaan
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Brianna J. Close
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Alexander H. Tavares
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Scott Seitz
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Nazimuddin Khan
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Sebastian Kapell
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| | - Nicholas A. Crossland
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Jonathan Z. Li
- Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Cambridge, MA, USA
| | - Florian Douam
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Susan C. Baker
- Department of Microbiology and Immunology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - John H. Connor
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
- Department of Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
| | - Mohsan Saeed
- Department of Biochemistry, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, USA
| |
Collapse
|
2
|
Rasmussen HB, Thomsen R, Hansen PR. Nucleoside analog GS-441524: pharmacokinetics in different species, safety, and potential effectiveness against Covid-19. Pharmacol Res Perspect 2022; 10:e00945. [PMID: 35396928 PMCID: PMC8994193 DOI: 10.1002/prp2.945] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 02/12/2022] [Indexed: 12/28/2022] Open
Abstract
GS-441524, the parent nucleoside of remdesivir, has been proposed to be effective against Covid-19 based on in vitro studies and studies in animals. However, randomized clinical trials of the agent to treat Covid-19 have not been conducted. Here, we evaluated GS-441524 for Covid-19 treatment based on studies reporting pharmacokinetic parameters of the agent in mice, rats, cats, dogs, monkeys, and the single individual in the first-in-human trial supplemented with information about its activity against severe acute respiratory syndrome coronavirus 2 and safety. A dosing interval of 8 h was considered clinically relevant and used to calculate steady-state plasma concentrations of GS-441524. These ranged from 0.27 to 234.41 μM, reflecting differences in species, doses, and administration routes. Fifty percent maximal inhibitory concentrations of GS-441524 against severe acute respiratory syndrome coronavirus 2 ranged from 0.08 μM to above 10 μM with a median of 0.87 μM whereas concentrations required to produce 90% of the maximal inhibition of the virus varied from 0.18 µM to more than 20 µM with a median of 1.42 µM in the collected data. Most of these concentrations were substantially lower than the calculated steady-state plasma concentrations of the agent. Plasma exposures to orally administered GS-441524, calculated after normalization of doses, were larger for dogs, mice, and rats than cynomolgus monkeys and humans, probably reflecting interspecies differences in oral uptake with reported oral bioavailabilities below 8.0% in cynomolgus monkeys and values as high as 92% in dogs. Reported oral bioavailabilities in rodents ranged from 12% to 57%. Using different presumptions, we estimated human oral bioavailability of GS-441524 at 13% and 20%. Importantly, doses of GS-441524 lower than the 13 mg/kg dose used in the first-in-human trial may be effective against Covid-19. Also, GS-441524 appears to be well-tolerated. In conclusion, GS-441524 has potential for oral treatment of Covid-19.
Collapse
Affiliation(s)
- Henrik Berg Rasmussen
- Institute of Biological Psychiatry, Mental Health Centre Sct. Hans, Roskilde, Denmark.,Department of Science and Environment, Roskilde University Center, Roskilde, Denmark
| | - Ragnar Thomsen
- Section of Forensic Chemistry, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter Riis Hansen
- Department of Cardiology, Herlev and Gentofte Hospital, Hellerup, Denmark
| |
Collapse
|
3
|
Groiss S, Somvilla I, Daxböck C, Fuchs J, Lang-Olip I, Stiegler P, Leber B, Liegl-Atzwanger B, Brislinger D. Quantification of increased MUC5AC expression in airway mucus of smoker using an automated image-based approach. Microsc Res Tech 2021; 85:5-18. [PMID: 34288207 DOI: 10.1002/jemt.23879] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
Microscopic analysis of mucus quantity and composition is crucial in research and diagnostics on muco-obstructive diseases. Currently used image-based methods are unable to extract concrete numeric values of mucosal proteins, especially on the expression of the key mucosal proteins MUC5AC and MUC5B. Since their levels increase under pathologic conditions such as extensive exposure to cigarette smoke, it is imperative to quantify them to improve treatment strategies of pulmonary diseases. This study presents a simple, image-based, and high-processing computational method that allows determining the ratio of MUC5AC and MUC5B within the overall airway mucus while providing information on their spatial distribution. The presented pipeline was optimized for automated downstream analysis using a combination of bright field and immunofluorescence imaging suitable for tracheal and bronchial tissue samples, and air-liquid interface (ALI) cell cultures. To validate our approach, we compared tracheal tissue and ALI cell cultures of isolated primary normal human bronchial epithelial cells derived from smokers and nonsmokers. Our data indicated 18-fold higher levels of MUC5AC in submucosal glands of smokers covering about 8% of mucosal areas compared to <1% in nonsmoking individuals, confirming results of previous studies. We further identified a subpopulation of nonsmokers with slightly elevated glandular MUC5AC levels suggesting moderate exposure to second-hand smoke or fine particulate air pollution. Overall, this study demonstrates a novel, user-friendly and freely available tool for digital pathology and the analysis of therapeutic interventions tested in ALI cell cultures.
Collapse
Affiliation(s)
- Silvia Groiss
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ina Somvilla
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Christine Daxböck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Ingrid Lang-Olip
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Philipp Stiegler
- Division of Transplantation Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Bettina Leber
- Division of Transplantation Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Bernadette Liegl-Atzwanger
- Diagnostic and Research Institute of Pathology, Diagnostic and Research Center for Molecular for Molecular Biomedicine, Medical University Graz, Graz, Austria
| | - Dagmar Brislinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
4
|
Screening for Effects of Inhaled Nanoparticles in Cell Culture Models for Prolonged Exposure. NANOMATERIALS 2021; 11:nano11030606. [PMID: 33671010 PMCID: PMC7997552 DOI: 10.3390/nano11030606] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/24/2022]
Abstract
Respiratory exposure of humans to environmental and therapeutic nanoparticles repeatedly occurs at relatively low concentrations. To identify adverse effects of particle accumulation under realistic conditions, monocultures of Calu-3 and A549 cells and co-cultures of A549 and THP-1 macrophages in the air–liquid interphase culture were exposed repeatedly to 2 µg/cm2 20 nm and 200 nm polystyrene particles with different functionalization. Particle accumulation, transepithelial electrical resistance, dextran (3–70 kDa) uptake and proinflammatory cytokine secretion were determined over 28 days. Calu-3 cells showed constant particle uptake without any change in barrier function and cytokine release. A549 cells preferentially ingested amino- and not-functionalized particles combined with decreased endocytosis. Cytokine release was transiently increased upon exposure to all particles. Carboxyl-functionalized demonstrated higher uptake and higher cytokine release than the other particles in the A549/THP-1 co-cultures. The evaluated respiratory cells and co-cultures ingested different amounts and types of particles and caused small (partly transient) effects. The data suggest that the healthy cells can adapt to low doses of non-cytotoxic particles.
Collapse
|
5
|
Montefusco-Pereira CV, Carvalho-Wodarz CDS, Seeger J, Kloft C, Michelet R, Lehr CM. Decoding (patho-)physiology of the lung by advanced in vitro models for developing novel anti-infectives therapies. Drug Discov Today 2020; 26:148-163. [PMID: 33232842 DOI: 10.1016/j.drudis.2020.10.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 09/11/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
Advanced lung cell culture models provide physiologically-relevant and complex data for mathematical models to exploit host-pathogen responses during anti-infective drug testing.
Collapse
Affiliation(s)
- Carlos Victor Montefusco-Pereira
- Department of Pharmacy, Saarland University, Saarbruecken, Germany; Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | | | - Johanna Seeger
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Saarbruecken, Germany; Department of Pharmacy, Saarland University, Saarbruecken, Germany
| |
Collapse
|
6
|
Cei D, Doryab A, Lenz AG, Schröppel A, Mayer P, Burgstaller G, Nossa R, Ahluwalia A, Schmid O. Development of a dynamic in vitro stretch model of the alveolar interface with aerosol delivery. Biotechnol Bioeng 2020; 118:690-702. [PMID: 33058147 DOI: 10.1002/bit.27600] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 10/01/2020] [Accepted: 10/09/2020] [Indexed: 01/07/2023]
Abstract
We describe the engineering design, computational modeling, and empirical performance of a moving air-liquid interface (MALI) bioreactor for the study of aerosol deposition on cells cultured on an elastic, porous membrane which mimics both air-liquid interface exposure conditions and mechanoelastic motion of lung tissue during breathing. The device consists of two chambers separated by a cell layer cultured on a porous, flexible membrane. The lower (basolateral) chamber is perfused with cell culture medium simulating blood circulation. The upper (apical) chamber representing the air compartment of the lung is interfaced to an aerosol generator and a pressure actuation system. By cycling the pressure in the apical chamber between 0 and 7 kPa, the membrane can mimic the periodic mechanical strain of the alveolar wall. Focusing on the engineering aspects of the system, we show that membrane strain can be monitored by measuring changes in pressure resulting from the movement of media in the basolateral chamber. Moreover, liquid aerosol deposition at a high dose delivery rate (>1 µl cm-2 min-1 ) is highly efficient (ca. 51.5%) and can be accurately modeled using finite element methods. Finally, we show that lung epithelial cells can be mechanically stimulated under air-liquid interface and stretch-conditions without loss of viability. The MALI bioreactor could be used to study the effects of aerosol on alveolar cells cultured at the air-liquid interface in a biodynamic environment or for toxicological or therapeutic applications.
Collapse
Affiliation(s)
- Daniele Cei
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Department of Information Engineering, University of Pisa, Pisa, Italy.,Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Ali Doryab
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Anke-Gabriele Lenz
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Andreas Schröppel
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Paula Mayer
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Gerald Burgstaller
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| | - Roberta Nossa
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Arti Ahluwalia
- Research Center "E. Piaggio", University of Pisa, Pisa, Italy.,Department of Information Engineering, University of Pisa, Pisa, Italy
| | - Otmar Schmid
- Comprehensive Pneumology Center, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease, Helmholtz Zentrum Muenchen, Neuherberg, Germany
| |
Collapse
|
7
|
Rotoli BM, Barilli A, Visigalli R, Ferrari F, Frati C, Lagrasta CA, Di Lascia M, Riccardi B, Puccini P, Dall’Asta V. Characterization of ABC Transporters in EpiAirway™, a Cellular Model of Normal Human Bronchial Epithelium. Int J Mol Sci 2020; 21:ijms21093190. [PMID: 32366035 PMCID: PMC7247561 DOI: 10.3390/ijms21093190] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
The ATP-binding cassette (ABC) transporters P-glycoprotein (MDR1/ABCB1), multidrug resistance-associated protein 1 (MRP1/ABCC1), and breast cancer resistance protein (BCRP/ABCG2) play a crucial role in the translocation of a broad range of drugs; data about their expression and activity in lung tissue are controversial. Here, we address their expression, localization and function in EpiAirway™, a three-dimensional (3D)-model of human airways; Calu-3 cells, a representative in vitro model of bronchial epithelium, are used for comparison. Transporter expression has been evaluated with RT-qPCR and Western blot, the localization with immunocytochemistry, and the activity by measuring the apical-to-basolateral and basolateral-to-apical fluxes of specific substrates in the presence of inhibitors. EpiAirway™ and Calu-3 cells express high levels of MRP1 on the basolateral membrane, while they profoundly differ in terms of BCRP and MDR1: BCRP is detected in EpiAirway™, but not in Calu-3 cells, while MDR1 is expressed and functional only in fully-differentiated Calu-3; in EpiAirway™, MDR1 expression and activity are undetectable, consistently with the absence of the protein in specimens from human healthy bronchi. In summary, EpiAirway™ appears to be a promising tool to study the mechanisms of drug delivery in the bronchial epithelium and to clarify the role of ABC transporters in the modulation of the bioavailability of administered drugs.
Collapse
Affiliation(s)
- Bianca Maria Rotoli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125 Parma, Italy; (A.B.); (R.V.); (F.F.); (V.D.)
- Correspondence: ; Tel.: +39-0521-033785
| | - Amelia Barilli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125 Parma, Italy; (A.B.); (R.V.); (F.F.); (V.D.)
| | - Rossana Visigalli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125 Parma, Italy; (A.B.); (R.V.); (F.F.); (V.D.)
| | - Francesca Ferrari
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125 Parma, Italy; (A.B.); (R.V.); (F.F.); (V.D.)
| | - Caterina Frati
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (C.F.); (C.A.L.)
| | - Costanza Annamaria Lagrasta
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Via Gramsci, 14, 43126 Parma, Italy; (C.F.); (C.A.L.)
| | - Maria Di Lascia
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo Francesco Belloli, 43122 Parma, Italy; (M.D.L.); (B.R.); (P.P.)
| | - Benedetta Riccardi
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo Francesco Belloli, 43122 Parma, Italy; (M.D.L.); (B.R.); (P.P.)
| | - Paola Puccini
- Preclinical Pharmacokinetics, Biochemistry & Metabolism Dept., Chiesi Farmaceutici, Largo Francesco Belloli, 43122 Parma, Italy; (M.D.L.); (B.R.); (P.P.)
| | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Via Volturno, 39, 43125 Parma, Italy; (A.B.); (R.V.); (F.F.); (V.D.)
| |
Collapse
|
8
|
Min KA, Zhang X, Yu JY, Rosania GR. Computational approaches to analyse and predict small molecule transport and distribution at cellular and subcellular levels. Biopharm Drug Dispos 2013; 35:15-32. [PMID: 24218242 DOI: 10.1002/bdd.1879] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 10/15/2013] [Accepted: 11/01/2013] [Indexed: 12/31/2022]
Abstract
Quantitative structure-activity relationship (QSAR) studies and mechanistic mathematical modeling approaches have been independently employed for analysing and predicting the transport and distribution of small molecule chemical agents in living organisms. Both of these computational approaches have been useful for interpreting experiments measuring the transport properties of small molecule chemical agents, in vitro and in vivo. Nevertheless, mechanistic cell-based pharmacokinetic models have been especially useful to guide the design of experiments probing the molecular pathways underlying small molecule transport phenomena. Unlike QSAR models, mechanistic models can be integrated from microscopic to macroscopic levels, to analyse the spatiotemporal dynamics of small molecule chemical agents from intracellular organelles to whole organs, well beyond the experiments and training data sets upon which the models are based. Based on differential equations, mechanistic models can also be integrated with other differential equations-based systems biology models of biochemical networks or signaling pathways. Although the origin and evolution of mathematical modeling approaches aimed at predicting drug transport and distribution has occurred independently from systems biology, we propose that the incorporation of mechanistic cell-based computational models of drug transport and distribution into a systems biology modeling framework is a logical next step for the advancement of systems pharmacology research.
Collapse
Affiliation(s)
- Kyoung Ah Min
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | | | | |
Collapse
|