1
|
Lykkesfeldt J, Carr AC, Tveden-Nyborg P. The pharmacology of vitamin C. Pharmacol Rev 2025; 77:100043. [PMID: 39986139 DOI: 10.1016/j.pharmr.2025.100043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
Ascorbic acid, the reduced form of vitamin C, is a ubiquitous small carbohydrate. Despite decades of focused research, new metabolic functions of this universal electron donor are still being discovered and add to the complexity of our view of vitamin C in human health. Although praised as an unsurpassed water-soluble antioxidant in plasma and cells, the most interesting functions of vitamin C seem to be its roles as specific electron donor in numerous biological reactions ranging from the well-known hydroxylation of proline to cofactor for the epigenetic master regulators ten-eleven translocation enzymes and Jumonji domain-containing histone-lysine demethylases. Some of these functions may have important implications for disease prevention and treatment and have spiked renewed interest in, eg, vitamin C's potential in cancer therapy. Moreover, some fundamental pharmacokinetic properties of vitamin C remain to be established including if other mechanisms than passive diffusion governs the efflux of ascorbate anions from the cell. Taken together, there still seems to be much to learn about the pharmacology of vitamin C and its role in health and disease. This review explores new avenues of vitamin C and integrates our present knowledge of its pharmacology. SIGNIFICANCE STATEMENT: Vitamin C is involved in multiple biological reactions of which most are essential to human health. Hundreds of millions of people are considered deficient in vitamin C according to accepted guidelines, but little is known about the long-term consequences. Although the complexity of vitamin C's physiology and pharmacology has been widely disregarded in clinical studies for decades, it seems clear that a deeper understanding of particularly its pharmacology holds the key to unravel and possibly exploit the potential of vitamin C in disease prevention and therapy.
Collapse
Affiliation(s)
- Jens Lykkesfeldt
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Anitra C Carr
- Nutrition in Medicine Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Pernille Tveden-Nyborg
- Section of Biomedicine, Department of Veterinary and Animal Science, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Warnken Z, Trementozzi A, Martins PP, Parkeh J, Koleng JJ, Smyth HDC, Brunaugh A. Development of Low-Cost, Weight-Adjustable Clofazimine Mini-Tablets for Treatment of Tuberculosis in Pediatrics. Eur J Pharm Sci 2023; 187:106470. [PMID: 37207942 DOI: 10.1016/j.ejps.2023.106470] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/16/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
Clofazimine (CFZ) is an important component of the World Health Organization's (WHO) recommended all-oral drug regimen for treatment of multi-drug resistant tuberculosis (MDR-TB). However, the lack of a dividable oral dosage form has limited the use of the drug in pediatric populations, who may require lowering of the dose to reduce the likelihood of adverse drug events. In this study, pediatric-friendly CFZ mini-tablets were prepared from micronized powder via direct compression. Rapid disintegration and maximized dissolution in GI fluids was achieved using an iterative formulation design process. Pharmacokinetic (PK) parameters of the optimized mini-tablets were obtained in Sprague-Dawley rats and compared against an oral suspension of micronized CFZ particles to examine the effect of processing and formulation on the oral absorption of the drug. Differences in maximum concentration and area under the curve between the two formulations were non-significant at the highest dosing level tested. Variability between rats prevented bioequivalence from being determined according to guidelines outlined by the Food and Drug Administration (FDA). These studies provide an important proof-of-concept for an alternative, low-cost formulation and processing approach for the oral delivery of CFZ in manner that is suitable for children as young as 6 months of age.
Collapse
Affiliation(s)
- Zachary Warnken
- Via Therapeutics, 2409 University Ave, Austin, TX, USA, 78712
| | | | | | - Jagruti Parkeh
- Via Therapeutics, 2409 University Ave, Austin, TX, USA, 78712
| | - John J Koleng
- Via Therapeutics, 2409 University Ave, Austin, TX, USA, 78712
| | - Hugh D C Smyth
- Via Therapeutics, 2409 University Ave, Austin, TX, USA, 78712; University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, 2409 University Ave, Austin, TX, USA, 78712
| | - Ashlee Brunaugh
- Via Therapeutics, 2409 University Ave, Austin, TX, USA, 78712; University of Michigan, College of Pharmacy, Department of Pharmaceutical Sciences, 428 Church St, Ann Arbor, MI, USA, 48109.
| |
Collapse
|
3
|
Hann E, Malagu K, Stott A, Vater H. The importance of plasma protein and tissue binding in a drug discovery program to successfully deliver a preclinical candidate. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:163-214. [PMID: 35753715 DOI: 10.1016/bs.pmch.2022.04.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Plasma protein binding and tissue binding are arguably two of the most critical parameters that are measured as part of a drug discovery program since, according to the free drug hypothesis, it is the free drug that is responsible for both efficacy and toxicity. This chapter aims to deconstruct the role of plasma protein and tissue binding in drug discovery programs, and to consider the conclusion made by Pfizer and Genentech/Depomed a decade ago that optimising plasma protein binding as an independent parameter does not significantly influence efficacy. This chapter will also examine how binding metrics are applied in drug discovery programs and explore circumstances where optimising plasma protein or tissue binding can be an effective strategy to deliver a candidate molecule for preclinical development with an early indication of sufficient therapeutic index.
Collapse
Affiliation(s)
- Elizabeth Hann
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom.
| | - Karine Malagu
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Andrew Stott
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| | - Huw Vater
- Charles River Laboratories, Robinson Building, Chesterford Research Park, Saffron Walden, United Kingdom
| |
Collapse
|
4
|
The Membrane Electrical Potential and Intracellular pH as Factors Influencing Intracellular Ascorbate Concentration and Their Role in Cancer Treatment. Cells 2021; 10:cells10112964. [PMID: 34831187 PMCID: PMC8616305 DOI: 10.3390/cells10112964] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ascorbate is an important element of a variety of cellular processes including the control of reactive oxygen species levels. Since reactive oxygen species are implicated as a key factor in tumorigenesis and antitumor therapy, the injection of a large amount of ascorbate is considered beneficial in cancer therapy. Recent studies have shown that ascorbate can cross the plasma membrane through passive diffusion. In contrast to absorption by active transport, which is facilitated by transport proteins (SVCT1 and SVCT2). The passive diffusion of a weak acid across membranes depends on the electrostatic potential and the pH gradients. This has been used to construct a new theoretical model capable of providing steady-state ascorbate concentration in the intracellular space and evaluating the time needed to reach it. The main conclusion of the analysis is that the steady-state intracellular ascorbate concentration weakly depends on its serum concentration but requires days of exposure to saturate. Based on these findings, it can be hypothesized that extended oral ascorbate delivery is possibly more effective than a short intravenous infusion of high ascorbate quantities.
Collapse
|
5
|
Di L. An update on the importance of plasma protein binding in drug discovery and development. Expert Opin Drug Discov 2021; 16:1453-1465. [PMID: 34403271 DOI: 10.1080/17460441.2021.1961741] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Plasma protein binding (PPB) remains a controversial topic in drug discovery and development. Fraction unbound (fu) is a critical parameter that needs to be measured accurately, because it has significant impacts on the predictions of drug-drug interactions (DDI), estimations of therapeutic indices (TI), and developments of PK/PD relationships. However, it is generally not advisable to change PPB through structural modifications, because PPB on its own has little relevance for in vivo efficacy.Areas covered: PPB fundamentals are discussed including the three main classes of drug binding proteins (i.e., albumin, alpha1-acid glycoprotein, and lipoproteins) and their physicochemical properties, in vivo half-life, and synthesis rate. State-of-the-art methodologies for PPB are highlighted. Applications of PPB in drug discovery and development are presented.Expert opinion: PPB is an old topic in pharmacokinetics, but there are still many misconceptions. Improving the accuracy of PPB for highly bound compounds is an ongoing effort in the field with high priority. As the field continues to generate high quality data, the regulatory agencies will increase their confidence in our ability to accurately measure PPB of highly bound compounds, and experimental fu values below 0.01 will more likely be used for DDI predictions in the future.
Collapse
Affiliation(s)
- Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, US
| |
Collapse
|
6
|
Bednarczyk D. Passive Influx and Ion Trapping Are More Relevant to the Cellular Accumulation of Highly Permeable Low-Molecular-Weight Acidic Drugs than Is Organic Anion Transporter 2. Drug Metab Dispos 2021; 49:648-657. [PMID: 34031139 DOI: 10.1124/dmd.121.000425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/17/2021] [Indexed: 11/22/2022] Open
Abstract
Recently published work suggests that highly permeable low-molecular-weight (LMW) acidic drugs are transported by organic anion transporter 2 (OAT2). However, an asymmetric distribution of ionizable drugs in subcellular organelles where pH gradients are significant may occur in the presence of an inhibitor relative to its absence (e.g., lysosomal trapping). In the present study, OAT2-mediated transport of highly permeable LMW anions could not be demonstrated using OAT2 transfected cells, despite robust transport of the OAT2 substrate penciclovir. Moreover, a rifamycin SV (RifSV)-dependent reduction in the accumulation of highly permeable LMW anions previously observed in hepatocytes could be qualitatively reproduced using HepG2 cells and also in Madin-Darby canine kidney (MDCK) cells, which lack expression of OAT2. Neither HepG2 nor MDCK cells demonstrated meaningful penciclovir transport, nor was the cellular accumulation of the highly permeable LMW anions sensitive to competitive inhibition by the neutral OAT2 substrate penciclovir. Both cell lines, however, demonstrated sensitivity to the mitochondrial uncoupler p-trifluoromethoxy carbonyl cyanide phenyl hydrazone (FCCP) in a manner similar to RifSV. Furthermore, the transepithelial MDCK permeability of the highly permeable LMW anions was measured in the absence and presence of RifSV and FCCP at concentrations that reduced the cellular accumulation of anions. Neither inhibitor, nor the OAT2 inhibitor ketoprofen, reduced the transepithelial flux of the anions as would be anticipated for transported substrate inhibition. The findings presented here are aligned with cellular accumulation of highly permeable LMW anions being significantly determined by ion trapping sensitive to mitochondrial uncoupling, rather than the result of OAT2-mediated transport. SIGNIFICANCE STATEMENT: The manuscript illustrates that passive influx and ion trapping are more relevant to the cellular accumulation of highly permeable low-molecular-weight acidic drugs than is the previously proposed mechanism of OAT2-mediated transport. The outcome illustrated here highlights a rare, and perhaps previously not reported, observation of anionic drug trapping in a compartment sensitive to mitochondrial uncoupling (e.g., the mitochondrial matrix) that may be confused for transporter-mediated uptake.
Collapse
Affiliation(s)
- Dallas Bednarczyk
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| |
Collapse
|
7
|
Di L, Riccardi K, Tess D. Evolving approaches on measurements and applications of intracellular free drug concentration and Kp uu in drug discovery. Expert Opin Drug Metab Toxicol 2021; 17:733-746. [PMID: 34058926 DOI: 10.1080/17425255.2021.1935866] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Introduction: Intracellular-free drug concentration (Cu,cell) and unbound partition coefficient (Kpuu) are two important parameters to develop pharmacokinetic and pharmacodynamic relationships, predict drug-drug interaction potentials and estimate therapeutic indices.Area covered: Methods on measurements of Cu,cell, Kpuu, partition coefficient (Kp) and fraction unbound of cells (fuc) are discussed. Advantages and limitations of several fuc methods are reviewed. Applications highlighted here are bridging the potency gaps between biochemical and cell-based assays, in vitro hepatocyte assay to predict in vivo liver-to-plasma Kpuu, the role of Kpuu in prediction of hepatic clearance for enzyme- and transporter-mediated mechanisms using extended clearance equation, and structural attributes governing tissue Kpuu.Expert opinion: Cu,cell and Kpuu are of growing applications in drug discovery. Methods for measurements of these properties continue to evolve in order to achieve higher precision/accuracy and obtain more detailed information at the subcellular levels. Future directions of the field include the development of in vitro and in silico models to predict tissue Kpuu, direct measurement of free drug concentration in subcellular organelles, and further investigations into the critical elements governing cell and tissue Kpuu. Significant innovation is needed to advance this complex, but highly impactful and exciting area of science.
Collapse
Affiliation(s)
- Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA
| | - Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA.,Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA
| | - David Tess
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT, USA.,Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA
| |
Collapse
|
8
|
Sharifian Gh M. Recent Experimental Developments in Studying Passive Membrane Transport of Drug Molecules. Mol Pharm 2021; 18:2122-2141. [PMID: 33914545 DOI: 10.1021/acs.molpharmaceut.1c00009] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The ability to measure the passive membrane permeation of drug-like molecules is of fundamental biological and pharmaceutical importance. Of significance, passive diffusion across the cellular membranes plays an effective role in the delivery of many pharmaceutical agents to intracellular targets. Hence, approaches for quantitative measurement of membrane permeability have been the topics of research for decades, resulting in sophisticated biomimetic systems coupled with advanced techniques. In this review, recent developments in experimental approaches along with theoretical models for quantitative and real-time analysis of membrane transport of drug-like molecules through mimetic and living cell membranes are discussed. The focus is on time-resolved fluorescence-based, surface plasmon resonance, and second-harmonic light scattering approaches. The current understanding of how properties of the membrane and permeant affect the permeation process is discussed.
Collapse
Affiliation(s)
- Mohammad Sharifian Gh
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia 22908, United States
| |
Collapse
|
9
|
Abstract
The study of enzyme kinetics in drug metabolism involves assessment of rates of metabolism and inhibitory potencies over a suitable concentration range. In all but the very simplest in vitro system, these drug concentrations can be influenced by a variety of nonspecific binding reservoirs that can reduce the available concentration to the enzyme system(s) under investigation. As a consequence, the apparent kinetic parameters, such as Km or Ki, that are derived can deviate from the true values. There are a number of sources of these nonspecific binding depots or barriers, including membrane permeation and partitioning, plasma or serum protein binding, and incubational binding. In the latter case, this includes binding to the assay apparatus as well as biological depots, depending on the characteristics of the in vitro matrix being used. Given the wide array of subcellular, cellular, and recombinant enzyme systems utilized in drug metabolism, each of these has different components which can influence the free drug concentration. The physicochemical properties of the test compound are also paramount in determining the influential factors in any deviation between true and apparent kinetic behavior. This chapter describes the underlying mechanisms determining the free drug concentration in vitro and how these factors can be accounted for in drug metabolism studies, illustrated with case studies from the literature.
Collapse
Affiliation(s)
- Nigel J Waters
- Preclinical Development, Black Diamond Therapeutics, Cambridge, MA, USA
| | - R Scott Obach
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Groton, CT, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc, Groton, CT, USA
| |
Collapse
|
10
|
Determination of Fraction Unbound and Unbound Partition Coefficient to Estimate Intracellular Free Drug Concentration. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2021. [DOI: 10.1007/978-1-0716-1250-7_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
11
|
Mathew S, Tess D, Burchett W, Chang G, Woody N, Keefer C, Orozco C, Lin J, Jordan S, Yamazaki S, Jones R, Di L. Evaluation of Prediction Accuracy for Volume of Distribution in Rat and Human Using In Vitro, In Vivo, PBPK and QSAR Methods. J Pharm Sci 2020; 110:1799-1823. [PMID: 33338491 DOI: 10.1016/j.xphs.2020.12.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 10/22/2022]
Abstract
Volume of distribution at steady state (Vss) is an important pharmacokinetic parameter of a drug candidate. In this study, Vss prediction accuracy was evaluated by using: (1) seven methods for rat with 56 compounds, (2) four methods for human with 1276 compounds, and (3) four in vivo methods and three Kp (partition coefficient) scalar methods from scaling of three preclinical species with 125 compounds. The results showed that the global QSAR models outperformed the PBPK methods. Tissue fraction unbound (fu,t) method with adipose and muscle also provided high Vss prediction accuracy. Overall, the high performing methods for human Vss prediction are the global QSAR models, Øie-Tozer and equivalency methods from scaling of preclinical species, as well as PBPK methods with Kp scalar from preclinical species. Certain input parameter ranges rendered PBPK models inaccurate due to mass balance issues. These were addressed using appropriate theoretical limit checks. Prediction accuracy of tissue Kp were also examined. The fu,t method predicted Kp values more accurately than the PBPK methods for adipose, heart and muscle. All the methods overpredicted brain Kp and underpredicted liver Kp due to transporter effects. Successful Vss prediction involves strategic integration of in silico, in vitro and in vivo approaches.
Collapse
Affiliation(s)
- Shibin Mathew
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - David Tess
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Cambridge, MA 02139, USA
| | - Woodrow Burchett
- Early Clinical Development, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - George Chang
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Nathaniel Woody
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Christopher Keefer
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Christine Orozco
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Jian Lin
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Samantha Jordan
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Shinji Yamazaki
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Rhys Jones
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, San Diego, CA 92121, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA.
| |
Collapse
|
12
|
Grünewald TGP, Alonso M, Avnet S, Banito A, Burdach S, Cidre‐Aranaz F, Di Pompo G, Distel M, Dorado‐Garcia H, Garcia‐Castro J, González‐González L, Grigoriadis AE, Kasan M, Koelsche C, Krumbholz M, Lecanda F, Lemma S, Longo DL, Madrigal‐Esquivel C, Morales‐Molina Á, Musa J, Ohmura S, Ory B, Pereira‐Silva M, Perut F, Rodriguez R, Seeling C, Al Shaaili N, Shaabani S, Shiavone K, Sinha S, Tomazou EM, Trautmann M, Vela M, Versleijen‐Jonkers YMH, Visgauss J, Zalacain M, Schober SJ, Lissat A, English WR, Baldini N, Heymann D. Sarcoma treatment in the era of molecular medicine. EMBO Mol Med 2020; 12:e11131. [PMID: 33047515 PMCID: PMC7645378 DOI: 10.15252/emmm.201911131] [Citation(s) in RCA: 170] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Sarcomas are heterogeneous and clinically challenging soft tissue and bone cancers. Although constituting only 1% of all human malignancies, sarcomas represent the second most common type of solid tumors in children and adolescents and comprise an important group of secondary malignancies. More than 100 histological subtypes have been characterized to date, and many more are being discovered due to molecular profiling. Owing to their mostly aggressive biological behavior, relative rarity, and occurrence at virtually every anatomical site, many sarcoma subtypes are in particular difficult-to-treat categories. Current multimodal treatment concepts combine surgery, polychemotherapy (with/without local hyperthermia), irradiation, immunotherapy, and/or targeted therapeutics. Recent scientific advancements have enabled a more precise molecular characterization of sarcoma subtypes and revealed novel therapeutic targets and prognostic/predictive biomarkers. This review aims at providing a comprehensive overview of the latest advances in the molecular biology of sarcomas and their effects on clinical oncology; it is meant for a broad readership ranging from novices to experts in the field of sarcoma.
Collapse
Affiliation(s)
- Thomas GP Grünewald
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Division of Translational Pediatric Sarcoma ResearchGerman Cancer Research Center (DKFZ), Hopp Children's Cancer Center (KiTZ), German Cancer Consortium (DKTK)HeidelbergGermany
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Marta Alonso
- Program in Solid Tumors and BiomarkersFoundation for the Applied Medical ResearchUniversity of Navarra PamplonaPamplonaSpain
| | - Sofia Avnet
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Ana Banito
- Pediatric Soft Tissue Sarcoma Research GroupGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Stefan Burdach
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Florencia Cidre‐Aranaz
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | - Gemma Di Pompo
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | | | | | | | | | | | - Merve Kasan
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | | | - Fernando Lecanda
- Division of OncologyAdhesion and Metastasis LaboratoryCenter for Applied Medical ResearchUniversity of NavarraPamplonaSpain
| | - Silvia Lemma
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Dario L Longo
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | | | | | - Julian Musa
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Shunya Ohmura
- Max‐Eder Research Group for Pediatric Sarcoma BiologyInstitute of PathologyFaculty of MedicineLMU MunichMunichGermany
| | | | - Miguel Pereira‐Silva
- Department of Pharmaceutical TechnologyFaculty of PharmacyUniversity of CoimbraCoimbraPortugal
| | - Francesca Perut
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Rene Rodriguez
- Instituto de Investigación Sanitaria del Principado de AsturiasOviedoSpain
- CIBER en oncología (CIBERONC)MadridSpain
| | | | - Nada Al Shaaili
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Shabnam Shaabani
- Department of Drug DesignUniversity of GroningenGroningenThe Netherlands
| | - Kristina Shiavone
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Snehadri Sinha
- Department of Oral and Maxillofacial DiseasesUniversity of HelsinkiHelsinkiFinland
| | | | - Marcel Trautmann
- Division of Translational PathologyGerhard‐Domagk‐Institute of PathologyMünster University HospitalMünsterGermany
| | - Maria Vela
- Hospital La Paz Institute for Health Research (IdiPAZ)MadridSpain
| | | | | | - Marta Zalacain
- Institute of Biostructures and Bioimaging (IBB)Italian National Research Council (CNR)TurinItaly
| | - Sebastian J Schober
- Department of Pediatrics and Children's Cancer Research Center (CCRC)Technische Universität MünchenMunichGermany
| | - Andrej Lissat
- University Children′s Hospital Zurich – Eleonoren FoundationKanton ZürichZürichSwitzerland
| | - William R English
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Nicola Baldini
- Orthopedic Pathophysiology and Regenerative Medicine UnitIRCCS Istituto Ortopedico RizzoliBolognaItaly
- Department of Biomedical and Neuromotor SciencesUniversity of BolognaBolognaItaly
| | - Dominique Heymann
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
- Université de NantesInstitut de Cancérologie de l'OuestTumor Heterogeneity and Precision MedicineSaint‐HerblainFrance
| |
Collapse
|
13
|
Pan BS, Perera SA, Piesvaux JA, Presland JP, Schroeder GK, Cumming JN, Trotter BW, Altman MD, Buevich AV, Cash B, Cemerski S, Chang W, Chen Y, Dandliker PJ, Feng G, Haidle A, Henderson T, Jewell J, Kariv I, Knemeyer I, Kopinja J, Lacey BM, Laskey J, Lesburg CA, Liang R, Long BJ, Lu M, Ma Y, Minnihan EC, O’Donnell G, Otte R, Price L, Rakhilina L, Sauvagnat B, Sharma S, Tyagarajan S, Woo H, Wyss DF, Xu S, Bennett DJ, Addona GH. An orally available non-nucleotide STING agonist with antitumor
activity. Science 2020; 369:369/6506/eaba6098. [DOI: 10.1126/science.aba6098] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 07/13/2020] [Indexed: 12/16/2022]
Abstract
Pharmacological activation of the STING (stimulator of interferon
genes)–controlled innate immune pathway is a promising therapeutic strategy for
cancer. Here we report the identification of MSA-2, an orally available
non-nucleotide human STING agonist. In syngeneic mouse tumor models, subcutaneous
and oral MSA-2 regimens were well tolerated and stimulated interferon-β secretion
in tumors, induced tumor regression with durable antitumor immunity, and
synergized with anti–PD-1 therapy. Experimental and theoretical analyses showed
that MSA-2 exists as interconverting monomers and dimers in solution, but only
dimers bind and activate STING. This model was validated by using synthetic
covalent MSA-2 dimers, which were potent agonists. Cellular potency of MSA-2
increased upon extracellular acidification, which mimics the tumor
microenvironment. These properties appear to underpin the favorable activity and
tolerability profiles of effective systemic administration of MSA-2.
Collapse
Affiliation(s)
- Bo-Sheng Pan
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Samanthi A. Perera
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Jeremy P. Presland
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Jared N. Cumming
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - B. Wesley Trotter
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Michael D. Altman
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Alexei V. Buevich
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Brandon Cash
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Saso Cemerski
- Department of Discovery Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Wonsuk Chang
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Yiping Chen
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Peter J. Dandliker
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Guo Feng
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Andrew Haidle
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Timothy Henderson
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - James Jewell
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Ilona Kariv
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Ian Knemeyer
- Department of Pharmacokinetics, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Johnny Kopinja
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Brian M. Lacey
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jason Laskey
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Charles A. Lesburg
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Rui Liang
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Brian J. Long
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Min Lu
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Yanhong Ma
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Ellen C. Minnihan
- Department of Discovery Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Greg O’Donnell
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Ryan Otte
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Laura Price
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Larissa Rakhilina
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Berengere Sauvagnat
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sharad Sharma
- Department of Discovery Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sriram Tyagarajan
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Hyun Woo
- Department of Pharmacokinetics, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Daniel F. Wyss
- Department of Discovery Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Serena Xu
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - George H. Addona
- Department of Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|
14
|
Achour B, Al-Majdoub ZM, Rostami-Hodjegan A, Barber J. Mass Spectrometry of Human Transporters. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:223-247. [PMID: 32084322 DOI: 10.1146/annurev-anchem-091719-024553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Transporters are key to understanding how an individual will respond to a particular dose of a drug. Two patients with similar systemic concentrations may have quite different local concentrations of a drug at the required site. The transporter profile of any individual depends upon a variety of genetic and environmental factors, including genotype, age, and diet status. Robust models (virtual patients) are therefore required and these models are data hungry. Necessary data include quantitative transporter profiles at the relevant organ. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently the most powerful method available for obtaining this information. Challenges include sourcing the tissue, isolating the hydrophobic membrane-embedded transporter proteins, preparing the samples for MS (including proteolytic digestion), choosing appropriate quantification methodology, and optimizing the LC-MS/MS conditions. Great progress has been made with all of these, especially within the last few years, and is discussed here.
Collapse
Affiliation(s)
- Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
- Certara, Princeton, New Jersey 08540, USA
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
15
|
Przybyło M, Langner M. On the physiological and cellular homeostasis of ascorbate. Cell Mol Biol Lett 2020; 25:32. [PMID: 32514268 PMCID: PMC7257198 DOI: 10.1186/s11658-020-00223-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
Recent interest in the role of ascorbate in crucial metabolic processes is driven by the growing number of medical reports that show beneficial effects of ascorbate supplementation for maintaining general well-being and recovery from a variety of medical conditions. The effect of ascorbate on the local body environment highly depends on its local concentration; at low concentrations it can cause the reduction of reactive oxygen and facilitate activities of enzymes, while at high concentrations it generates free radicals by reducing ferric ions. Ascorbate serving as an electron donor assists the iron-containing proteins and the iron transfer between various aqueous compartments. These functions require effective and adjustable mechanisms responsible for ascorbate biodistribution. In the paper we propose a new biophysical model of ascorbate redistribution between various aqueous body compartments. It combines recent experimental evidence regarding the ability of ascorbate to cross the lipid bilayer by unassisted diffusion, with active transport by well-characterized sodium vitamin C transporter (SVCT) membrane proteins. In the model, the intracellular concentration of ascorbate is maintained by the balance of two opposing fluxes: fast active and slow passive transport. The model provides a mechanistic understanding of ascorbate flux across the epidermal barrier in the gut as well as the role of astrocytes in ascorbate recycling in the brain. In addition, ascorbate passive diffusion across biological membranes, which depends on membrane electric potentials and pH gradients, provides the rationale for the correlation between ascorbate distribution and the transfer of iron ions inside a cell. The proposed approach provides, for the first time, a mechanistic account of processes leading to ascorbate physiological and cellular distribution, which helps to explain numerous experimental and clinical observations.
Collapse
Affiliation(s)
- Magdalena Przybyło
- Faculty of Biomedical Engineering, Wrocław University of Sciences and Technology, 50-370 Wrocław, Poland
- Lipid Systems Ltd, Krzemieniecka 48C, 54-613 Wrocław, Poland
| | - Marek Langner
- Faculty of Biomedical Engineering, Wrocław University of Sciences and Technology, 50-370 Wrocław, Poland
- Lipid Systems Ltd, Krzemieniecka 48C, 54-613 Wrocław, Poland
| |
Collapse
|
16
|
Orozco CC, Atkinson K, Ryu S, Chang G, Keefer C, Lin J, Riccardi K, Mongillo RK, Tess D, Filipski KJ, Kalgutkar AS, Litchfield J, Scott D, Di L. Structural attributes influencing unbound tissue distribution. Eur J Med Chem 2020; 185:111813. [DOI: 10.1016/j.ejmech.2019.111813] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 12/26/2022]
|
17
|
Marzuoli I, Margreitter C, Fraternali F. Lipid Head Group Parameterization for GROMOS 54A8: A Consistent Approach with Protein Force Field Description. J Chem Theory Comput 2019; 15:5175-5193. [PMID: 31433640 PMCID: PMC7377650 DOI: 10.1021/acs.jctc.9b00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Membranes
are a crucial component of both bacterial and mammalian
cells, being involved in signaling, transport, and compartmentalization.
This versatility requires a variety of lipid species to tailor the
membrane’s behavior as needed, increasing the complexity of
the system. Molecular dynamics simulations have been successfully
applied to study model membranes and their interactions with proteins,
elucidating some crucial mechanisms at the atomistic detail and thus
complementing experimental techniques. An accurate description of
the functional interplay of the diverse membrane components crucially
depends on the selected parameters that define the adopted force field.
A coherent parameterization for lipids and proteins is therefore needed.
In this work, we propose and validate new lipid head group parameters
for the GROMOS 54A8 force field, making use of recently published
parametrizations for key chemical moieties present in lipids. We make
use additionally of a new canonical set of partial charges for lipids,
chosen to be consistent with the parameterization of soluble molecules
such as proteins. We test the derived parameters on five phosphocholine
model bilayers, composed of lipid patches four times larger than the
ones used in previous studies, and run 500 ns long simulations of
each system. Reproduction of experimental data like area per lipid
and deuterium order parameters is good and comparable with previous
parameterizations, as well as the description of liquid crystal to
gel-phase transition. On the other hand, the orientational behavior
of the head groups is more realistic for this new parameter set, and
this can be crucial in the description of interactions with other
polar molecules. For that reason, we tested the interaction of the
antimicrobial peptide lactoferricin with two model membranes showing
that the new parameters lead to a weaker peptide–membrane binding
and give a more realistic outcome in comparing binding to antimicrobial
versus mammal membranes.
Collapse
Affiliation(s)
- Irene Marzuoli
- Randall Centre for Cell and Molecular Biology , King's College London , London SE1 1UL , U.K
| | - Christian Margreitter
- Randall Centre for Cell and Molecular Biology , King's College London , London SE1 1UL , U.K
| | - Franca Fraternali
- Randall Centre for Cell and Molecular Biology , King's College London , London SE1 1UL , U.K
| |
Collapse
|
18
|
Russo G, Barbato F, Grumetto L, Philippe L, Lynen F, Goetz GH. Entry of therapeutics into the brain: Influence of exposed polarity calculated in silico and measured in vitro by supercritical fluid chromatography. Int J Pharm 2019; 560:294-305. [DOI: 10.1016/j.ijpharm.2019.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 01/18/2019] [Accepted: 02/08/2019] [Indexed: 12/23/2022]
|
19
|
Fathizadeh A, Elber R. Ion Permeation through a Phospholipid Membrane: Transition State, Path Splitting, and Calculation of Permeability. J Chem Theory Comput 2019; 15:720-730. [PMID: 30474968 PMCID: PMC6467798 DOI: 10.1021/acs.jctc.8b00882] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigate the thermodynamics and kinetics of the permeation of a potassium ion through a phospholipid membrane. We illustrate that the conventional reaction coordinate (the position of the ion along the normal to the membrane plane) is insufficient to capture essential elements of the process. It is necessary to add coarse variables that measure membrane distortion. New coarse variables are suggested, and a two-dimensional coarse-space is proposed to describe the permeation. We illustrate path splitting and two transition states of comparable barrier heights. The alternative pathways differ by the extent of water solvation of the ion-phosphate pairs. The permeation process cannot be described by a local one-dimensional reaction coordinate, and a network formulation is more appropriate. We use Milestoning with Voronoi tessellation in two dimensions to quantify the equilibrium and rate of the permeation of the positively charged ion. The permeation coefficient is computed and compared favorably to experiment.
Collapse
Affiliation(s)
- Arman Fathizadeh
- Institute for Computational Engineering and Sciences , University of Texas at Austin , Austin , Texas 78712 , United States
| | - Ron Elber
- Institute for Computational Engineering and Sciences , University of Texas at Austin , Austin , Texas 78712 , United States
- Department of Chemistry , University of Texas at Austin , Austin , Texas 78712 , United States
| |
Collapse
|
20
|
Zhu Z, Li Y, Yang X, Pan W, Pan H. The reversion of anti-cancer drug antagonism of tamoxifen and docetaxel by the hyaluronic acid-decorated polymeric nanoparticles. Pharmacol Res 2017; 126:84-96. [PMID: 28734999 DOI: 10.1016/j.phrs.2017.07.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/24/2017] [Accepted: 07/11/2017] [Indexed: 12/11/2022]
Abstract
Docetaxel (DTX) and tamoxifen (TMX) are first-line drugs used to treat breast cancer. However when used in combination, they produce antagonism because of their differential metabolic pathways. In order to prevent this antagonism, an amphiphilic copolymer, cholesterol modified hyaruronic acid (HA-CHOL), was synthesized for investigating the co-delivery of TMX and DTX. In vitro drug release experiment of the Co-encapsulated (encapsulated DTX+TMX) nanoparticles (Co-NPs) revealed that NPs with unique release mechanism can markedly reduce the release of these drugs in the circulatory system. However, when reaching in cell, TMX can release rapidly to prevent DTX from coming into contact with metabolizing enzymes. In vitro cytotoxicity experiment revealed that the Co-NPs exhibited a significant synergistic effect for inhibiting the proliferation of the cancer cell lines A549, MCF7 and S180. NPs carrying Coumarin-6(Cou6) exhibited increased cellular uptake compared with Cou6 solution at similar drug concentrations. As an in vivo treatment of xenograft tumors involving 180 cells, the Co-NPs displayed a clear tumor-inhibiting effect. This led us to conclude that the reversion of drug antagonism by NPs was attributed to the increased stability of the nanoparticles in the blood circulation, the efficient cellular uptake, the hierarchical drug metabolism in the tumor and the good and orderly delivery of the drugs to the tumor tissue.
Collapse
Affiliation(s)
- Zhihong Zhu
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Yuenan Li
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Xinggang Yang
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China
| | - Weisan Pan
- School of Pharmacy, Shenyang Pharmaceutics University, Shenyang, Liaoning, 110016, China, China.
| | - Hao Pan
- College of Pharmacy, Liaoning University, Shenyang, 110036, China, China.
| |
Collapse
|
21
|
Riccardi K, Lin J, Li Z, Niosi M, Ryu S, Hua W, Atkinson K, Kosa RE, Litchfield J, Di L. Novel Method to Predict In Vivo Liver-to-Plasma K puu for OATP Substrates Using Suspension Hepatocytes. Drug Metab Dispos 2017; 45:576-580. [PMID: 28258068 DOI: 10.1124/dmd.116.074575] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/01/2017] [Indexed: 02/13/2025] Open
Abstract
The ability to predict human liver-to-plasma unbound partition coefficient (Kpuu) is of great importance to estimate unbound liver concentration, develop PK/PD relationships, predict efficacy and toxicity in the liver, and model the drug-drug interaction potential for drugs that are asymmetrically distributed into the liver. A novel in vitro method has been developed to predict in vivo Kpuu with good accuracy using cryopreserved suspension hepatocytes in InVitroGRO HI media with 4% BSA. Validation was performed using six OATP substrates with rat in vivo Kpuu data from i.v. infusion studies where a steady state was achieved. Good in vitro-in vivo correlation (IVIVE) was observed as the in vitro Kpuu values were mostly within 2-fold of in vivo Kpuu Good Kpuu IVIVE in human was also observed with in vivo Kpuu data of dehydropravastatin from positron emission tomography and in vivo Kpuu data from PK/PD modeling for pravastatin and rosuvastatin. Under the specific Kpuu assay conditions, the drug-metabolizing enzymes and influx/efflux transporters appear to function at physiologic levels. No scaling factors are necessary to predict in vivo Kpuu from in vitro data. The novel in vitro Kpuu method provides a useful tool in drug discovery to project in vivo Kpuu.
Collapse
Affiliation(s)
- Keith Riccardi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Jian Lin
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Zhenhong Li
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Mark Niosi
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Sangwoo Ryu
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Wenyi Hua
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Karen Atkinson
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Rachel E Kosa
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - John Litchfield
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Inc., Groton, Connecticut (K.R., Ji.L., M.N., S.R., W.H., K.A., R.E.K., L.D.); Cambridge, Massachusetts (Z.L., Jo.L.)
| |
Collapse
|