1
|
Timmerman P, White S, Adcock N, Arfvidsson C, Barfield M, Cowan K, Ferrari L, Golob M, Goodwin L, Hughes R, Ivanova T, Laurén A, McDougall S, Nelson R, van de Merbel N, Verhaeghe T, Wright M. Feedback from a workshop by the European Bioanalysis Forum on assay validation requirements for in vitro assays following the publication of ICH M12 guideline - a plea for context-of-use over ICH M10 standards. Bioanalysis 2025; 17:379-382. [PMID: 39985138 PMCID: PMC11959917 DOI: 10.1080/17576180.2025.2468596] [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: 01/24/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025] Open
Abstract
The release of the ICH M12 Guideline on Drug Interaction Studies has reignited discussions around assay validation requirements for in vitro assays such as plasma protein-binding studies. Even though the ICH M12 does not directly reference the ICH M10 Guideline on Bioanalytical Method Validation and Sample Analysis, its release prompted further discussions on assay validation requirements for these studies during the 17th European Bioanalysis Forum Open Symposium held in Barcelona, Spain, from 19 to 21 November 2024, where we advocated for a Context-of-Use driven approach over rigid adherence to ICH M10 standards. Context-of-Use driven validation ensures assays are tailored to the specific scientific and regulatory needs, optimizing resource allocation and innovation in drug development. This short opinion paper explores the potential and undesired implications of ICH M12 on bioanalytical validation practices, highlights the distinction between exploratory assays and assays having a clinical impact, and underscores the necessity for tailored validation strategies.
Collapse
Affiliation(s)
| | - Steve White
- GSK, Precision Medicine Design Assurance, Stevenage, UK
| | | | - Cecilia Arfvidsson
- AstraZeneca, Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, R&D, Gothenburg, Sweden
| | - Matthew Barfield
- Roche Innovation Center Welwyn, Roche Pharma Research and Early Development, Welwyn Garden City, UK
| | - Kyra Cowan
- Merck KGaA, Research and Development, Drug Metabolism and Pharmacokinetics New Biological Entities, Darmstadt, Germany
| | - Luca Ferrari
- Roche Pharma Research & Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Lee Goodwin
- Labcorp, Bioanalytical Services, Harrogate, UK
| | | | | | | | - Stuart McDougall
- Quotient Sciences, Bioanalytical Department, Alnwick, Northumberland, UK
| | | | | | | | - Michael Wright
- GSK, Biomarker & Bioanalytical Platforms, Precision Medicine, Stevenage, UK
| |
Collapse
|
2
|
Li L, Ngougni Pokem P, Sassen SDT, Wittebole X, Laterre PF, Vervaeke S, Zeitlinger M, Van Bambeke F, Muller AE. Variable temocillin protein binding and pharmacokinetics in different clinical conditions: Implications for target attainment. Br J Clin Pharmacol 2025. [PMID: 39840787 DOI: 10.1111/bcp.16397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/01/2024] [Accepted: 12/25/2024] [Indexed: 01/23/2025] Open
Abstract
AIMS The beta-lactam antibiotic temocillin is increasingly used to treat extended-spectrum beta-lactamase (ESBL-producing) strains; however, its protein binding is complex. This study aims to predict unbound temocillin concentrations in various participant groups to determine its impact on the probability of target attainment (PTA) and to improve dosing recommendations. METHODS The plasma pharmacokinetics were analysed using non-linear mixed-effects modelling. Data from individuals in four groups: healthy volunteers (HV), urinary tract infection patients (UTI), ventriculitis patients and sepsis-ICU patients were included. Simulations were performed to compare the PTA for different dosing regimens and participant-groups. RESULTS A two-compartment protein-binding model best fitted the 1085 concentrations (543 unbound, 542 total). Temocillin clearance was influenced by creatinine clearance, serum albumin (ALB) and C-reactive protein (CRP). For 2 g q8h intermittent infusion, the PTAs at an MIC of 16 mg/L were 2.3%, 39.5%, 10.0% and 72.5%, for HV, UTI, ventriculitis and sepsis-ICU patients, respectively. The effects of the covariates on the PTA were simulated for two example patients with intermittent infusion: the PTAs at an MIC of 8 mg/L for a sepsis-ICU patient (CRP 300 mg/L, albumin 15 g/L) and a mild-UTI patient (CRP 30 mg/L, albumin 35 g/L) were 94.3% and 62.4%, respectively. Continuous infusion consistently outperformed intermittent infusion in achieving the desired pharmacodynamic target (time above MIC). CONCLUSIONS Our study underscores the significant variation in temocillin clearance and unbound fractions among different participant groups, challenging the efficacy of traditional 2 g q12h dosing. For patients with enhanced renal function and lower inflammation, continuous infusion emerges as a more effective strategy to achieve optimal target attainment.
Collapse
Affiliation(s)
- Letao Li
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Perrin Ngougni Pokem
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Sebastiaan D T Sassen
- Department of Hospital Pharmacy, Erasmus University Medical Center, Rotterdam, The Netherlands
- CATOR, Center for Antimicrobial Optimized Treatment-Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics Group, Rotterdam, The Netherlands
| | - Xavier Wittebole
- Department of Critical Care Medicine, Cliniques universitaires St Luc, UCLouvain, Brussels, Belgium
| | - Pierre François Laterre
- Department of Critical Care Medicine, Cliniques universitaires St Luc, UCLouvain, Brussels, Belgium
- Department of Critical Care Medicine, CHR Mons-Hainaut, Mons, Belgium
| | - Steven Vervaeke
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Anouk E Muller
- Department of Medical Microbiology, Haaglanden Medisch Centrum, The Hague, The Netherlands
- CATOR, Center for Antimicrobial Optimized Treatment-Rotterdam, Rotterdam, The Netherlands
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Lederberg OL, Yan NL, Sanchez J, Ren W, Ash C, Wilkens SJ, Qiu H, Qin B, Grant VH, Jackman AB, Stanfield RL, Wilson IA, Petrassi HM, Rhoades D, Kelly JW. Discovery of Potent and Selective Pyridone-Based Small Molecule Kinetic Stabilizers of Amyloidogenic Immunoglobulin Light Chains. J Med Chem 2024; 67:21070-21105. [PMID: 39626211 DOI: 10.1021/acs.jmedchem.4c01773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Kinetic stabilization of amyloidogenic immunoglobulin light chains (LCs) through small molecule binding may become the first treatment for the proteinopathy component of light chain amyloidosis (AL). Kinetic stabilizers selectively bind to the native state over the misfolding transition state, slowing denaturation. Prior λ full-length LC dimer (FL LC2) kinetic stabilizers exhibited considerable plasma protein binding. We hypothesized that the coumarin "aromatic core" of the stabilizers was responsible for the undesirable plasma protein binding. Here, we describe structure-activity relationship (SAR) data initially focused on replacing the coumarin aromatic core. 2-pyridones proved suitable replacements. We subsequently optimized the "anchor substructure" in the context of 2-pyridones, resulting in potent λ FL LC2 kinetic stabilizers exhibiting reduced plasma protein binding. The 3-methyl- or 3-ethyl-3-phenylpyrrolidine-2-pyridone scaffold stabilized multiple AL patient-derived λ FL LC2s in human plasma. This, coupled with X-ray crystallographic data, indicates that 3-alkyl-3-phenylpyrrolidine-2-pyridone-based stabilizers are promising candidates for treating the proteinopathy component of AL.
Collapse
Affiliation(s)
- Oren L Lederberg
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Nicholas L Yan
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Julian Sanchez
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Wen Ren
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Carl Ash
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Steven J Wilkens
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Huang Qiu
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Bo Qin
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Virginia H Grant
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Alex B Jackman
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road., La Jolla, California 92037, United States
| | - H Michael Petrassi
- Protego Biopharma, 10945 Vista Sorrento Parkway, San Diego, California 92130, United States
| | - Derek Rhoades
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road., La Jolla, California 92037, United States
| |
Collapse
|
4
|
Martinelli E, Spiller M, Weck R, Llompart P, Minoletti C, Güssregen S, Sib A, Derdau V. Pegylated Phosphine Ligands in Iridium(I) Catalyzed Hydrogen Isotope Exchange Reactions in Aqueous Buffers. Chemistry 2024; 30:e202402038. [PMID: 38861127 DOI: 10.1002/chem.202402038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/12/2024]
Abstract
The synthesis of a water-soluble, phosphine-pegylated iridium(I) catalyst and its application in hydrogen isotope exchange (HIE) reactions in buffer is reported. The longer polyethylene glycol side chains on the phosphine increased the water solubility independently from the pH. HIE reactions of polar substrates in protic solvents were studied. DFT calculations gave further insights into the catalytic processes. The scope and limitation of the pegylated catalyst was studied in HIE reactions of several complex compounds in borax buffer at pH 9 and the best conditions were applied in a tritium experiment with the drug telmisartan.
Collapse
Affiliation(s)
- Elisa Martinelli
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| | - Marie Spiller
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| | - Remo Weck
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| | - Pierre Llompart
- Sanofi France, Integrated Drug Discovery, 1 impasse des Ateliers, 94 400, Vitry-sur-Seine, France
| | - Claire Minoletti
- Sanofi France, Integrated Drug Discovery, 1 impasse des Ateliers, 94 400, Vitry-sur-Seine, France
| | - Stefan Güssregen
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| | - Anna Sib
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| | - Volker Derdau
- Sanofi Germany, Integrated Drug Discovery, Industriepark Höchst, G876, 65926, Frankfurt am Main, Germany
| |
Collapse
|
5
|
Lier C, Dejaco A, Kratzer A, Kees MG, Kees F, Dorn C. Free serum concentrations of antibiotics determined by ultrafiltration: extensive evaluation of experimental variables. Bioanalysis 2024; 16:747-756. [PMID: 39041640 PMCID: PMC11389746 DOI: 10.1080/17576180.2024.2365526] [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: 03/12/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024] Open
Abstract
Aim: To assess the impact of experimental conditions on free serum concentrations as determined by ultrafiltration and HPLC-DAD analysis in a wide range of antibiotics.Materials & methods: Relative centrifugation force (RCF), temperature, pH and buffer were varied and the results compared with the standard protocol (phosphate buffer pH 7.4, 37°C, 1000 × g).Results: Generally, at 10,000 × g the unbound fraction (fu) decreased with increasing molecular weight, and was lower at 22°C. In unbuffered serum, the fu of flucloxacillin or valproic acid was increased, that of basic or amphoteric drugs considerably decreased. Comparable results were obtained using phosphate or HEPES buffer except for drugs which form metal chelate complexes.Conclusion: Maintaining a physiological pH is more important than strictly maintaining body temperature.
Collapse
Affiliation(s)
- Constantin Lier
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Alexander Dejaco
- Department of Anaesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Kratzer
- Hospital Pharmacy, University Hospital Regensburg, Regensburg, Germany
| | - Martin G Kees
- Department of Anaesthesiology, University Hospital Regensburg, Regensburg, Germany
| | - Frieder Kees
- Department of Pharmacology, University of Regensburg, Regensburg, Germany
| | - Christoph Dorn
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
6
|
Manhas D, Dhiman S, Kour H, Kour D, Sharma K, Wazir P, Vij B, Kumar A, Sawant SD, Ahmed Z, Nandi U. ADME/PK Insights of Crocetin: A Molecule Having an Unusual Chemical Structure with Druglike Features. ACS OMEGA 2024; 9:21494-21509. [PMID: 38764638 PMCID: PMC11097163 DOI: 10.1021/acsomega.4c02116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 05/21/2024]
Abstract
Crocetin is a promising phyto-based molecule to treat Alzheimer's disease (AD). The chemical structure of crocetin is incongruent with various standard structural features of CNS drugs. As poor pharmacokinetic behavior is the major hurdle for any candidate to become a drug, we elucidated its druggable characteristics by implementing in silico, in vitro, and in vivo approaches, as limited ADME/PK information is available. Results demonstrate several attributes of crocetin based on rules of drug-likeness, lipophilicity, pKa, P-gp inhibitory activity, plasma stability, RBC partitioning, metabolic stability, CYP inhibitory action, blood-brain barrier (BBB) permeability, oral bioavailability, and pharmacokinetic interaction with marketed anti-Alzheimer's drugs (memantine, donepezil, galantamine, and rivastigmine). However, aqueous solubility, chemical stability, plasma protein binding, and P-gp induction are some concerns associated with this molecule that should be taken into consideration during its further development. Overall results indicate favorable ADME/PK behavior and potential druggable candidature of crocetin.
Collapse
Affiliation(s)
- Diksha Manhas
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit Dhiman
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harpreet Kour
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Dilpreet Kour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuhu Sharma
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priya Wazir
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
| | - Bhavna Vij
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
| | - Ajay Kumar
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanghapal D. Sawant
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Zabeer Ahmed
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
7
|
Woziński M, Greber KE, Pastewska M, Kolasiński P, Hewelt-Belka W, Żołnowska B, Sławiński J, Szulczyk D, Sawicki W, Ciura K. Modification of gradient HPLC method for determination of small molecules' affinity to human serum albumin under column safety conditions: Robustness and chemometrics study. J Pharm Biomed Anal 2024; 239:115916. [PMID: 38134704 DOI: 10.1016/j.jpba.2023.115916] [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: 10/16/2023] [Revised: 11/19/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023]
Abstract
In the early stages of drug discovery, beyond the biological activity screening, determining the physicochemical properties that affect the distribution of molecules in the human body is an essential step. Plasma protein binding (PPB) is one of the most important investigated endpoints. Nevertheless, the methodology for measuring %PPB is significantly less popular and standardized than other physicochemical properties, like lipophilicity. Here, we proposed how to modify protocols presented by Valko into column safety conditions and evaluated their robustness using fractional factorial design. For robustness testing, four factors were selected: column temperature, mobile phase flow rate, maximum isopropanol concentration in the mobile phase, and buffer pH. Elaborate methods have been applied for the analysis of HSA affinity for three groups of antibiotic-oriented substances that vary in chemical structure: fluoroquinolones, sulfonamides, and tetrazole derivatives. Furthermore, based on the reversed-phase chromatography the workflow of pilot studies was proposed to select molecules that have high affinity to HSA and can not be eluted from the HSA column using the concentration of organic modifier recommended by the column manufacturer.
Collapse
Affiliation(s)
- Mateusz Woziński
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Katarzyna Ewa Greber
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Monika Pastewska
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Piotr Kolasiński
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Weronika Hewelt-Belka
- Department of Analytical Chemistry, Chemical Faculty, Gdańsk University of Technology, G. Narutowicza 11/12, 80-233 Gdańsk, Poland
| | - Beata Żołnowska
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Jarosław Sławiński
- Department of Organic Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Wiesław Sawicki
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland
| | - Krzesimir Ciura
- Department of Physical Chemistry, Medical University of Gdańsk, Al. Gen. Hallera 107, 80-416 Gdańsk, Poland; QSAR Lab Ltd., Trzy Lipy 3 St. Gdańsk, 80-172, Poland.
| |
Collapse
|
8
|
Wen A, Qin ARR, Tarnowski T, Ling KHJ, Zhang H, Humeniuk R, Regan S, Saquing J, Liu W, Venkatarangan L, Xiao D. Plasma Protein Binding Determination for Unstable Ester Prodrugs: Remdesivir and Tenofovir Alafenamide. J Pharm Sci 2023; 112:3224-3232. [PMID: 37722451 DOI: 10.1016/j.xphs.2023.09.009] [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: 06/22/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Remdesivir (RDV) and tenofovir alafenamide (TAF) are prodrugs designed to be converted to their respective active metabolites. Plasma protein binding (PPB) determination of these prodrugs is important for patients with possible alteration of free fraction of the drugs due to plasma protein changes in renal impairment, hepatic impairment, or pregnancy. However, the prodrugs' instability in human plasma presents a challenge for accurate PPB determination. In this research work, two approaches were used in the method development and qualification for PPB assessment of RDV and TAF. For RDV, dichlorvos was used to inhibit esterase activity to stabilize the prodrug in plasma during equilibrium dialysis (ED). The impact of dichlorvos on protein binding was evaluated and determined to be insignificant by comparing the unbound fraction (fu) determined by the ED method with dichlorvos present and the fu determined by an ultrafiltration method without dichlorvos. In contrast to RDV, TAF degradation in plasma is ∼3-fold slower, and TAF stability cannot be improved by dichlorvos. Fit-for-purpose acceptance criteria for the TAF PPB method were chosen, and an ED method was developed based on these criteria. These two methods were then qualified and applied for PPB determinations in clinical studies.
Collapse
Affiliation(s)
- Anita Wen
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Ann Ran-Ran Qin
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Thomas Tarnowski
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | | | - Haeyoung Zhang
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Rita Humeniuk
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Sean Regan
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | | | | | | | - Deqing Xiao
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA.
| |
Collapse
|
9
|
Kramp H, Weck R, Sandvoss M, Sib A, Mencia G, Fazzini PF, Chaudret B, Derdau V. In situ Generated Iridium Nanoparticles as Hydride Donors in Photoredox-Catalyzed Hydrogen Isotope Exchange Reactions with Deuterium and Tritium Gas. Angew Chem Int Ed Engl 2023; 62:e202308983. [PMID: 37453077 DOI: 10.1002/anie.202308983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023]
Abstract
We have studied the photoredox-catalyzed hydrogen isotope exchange (HIE) reaction with deuterium or tritium gas as isotope sources and in situ formed transition metal nanoparticles as hydrogen atom transfer pre-catalysts. By this means we have found synergistic reactivities applying two different HIE mechanisms, namely photoredox-catalyzed and CH-functionalization HIE leading to the synthesis of highly deuterated complex molecules. Finally, we adopted these findings successfully to tritium chemistry.
Collapse
Affiliation(s)
- Henrik Kramp
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Remo Weck
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Martin Sandvoss
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Anna Sib
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| | - Gabriel Mencia
- Laboratoire de Physique et Chimie des Nano-objets, Institut National des Sciences Appliquées, 135 avenue de Rangueil, 31077, Toulouse Cedex 4, France
| | - Pier-Francesco Fazzini
- Laboratoire de Physique et Chimie des Nano-objets, Institut National des Sciences Appliquées, 135 avenue de Rangueil, 31077, Toulouse Cedex 4, France
| | - Bruno Chaudret
- Laboratoire de Physique et Chimie des Nano-objets, Institut National des Sciences Appliquées, 135 avenue de Rangueil, 31077, Toulouse Cedex 4, France
| | - Volker Derdau
- Sanofi Germany, R&D, Integrated Drug Discovery, Industriepark Höchst, 65926, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Wientjes MG, Lu Z, Chan CHF, Turaga K, Au JLS. Surgical management of peritoneal metastasis: Opportunities for pharmaceutical research. J Control Release 2023; 361:717-726. [PMID: 37574051 PMCID: PMC10560040 DOI: 10.1016/j.jconrel.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/20/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Cytoreductive surgery (CRS) has emerged as a survival-extending treatment of peritoneal metastasis (PM); recent advances include using intraperitoneal chemotherapy (IPC) at normothermic or hyperthermic temperatures, or under pressure (CRS + IPC). Clinical CRS + IPC research has established its highly variable efficacy and suggested tumor size, tumor locations and presence of ascites as potential determinants. On the other hand, there is limited knowledge on the effects of pharmaceutical properties on treatment outcomes. The present study investigated the inter-subject variability of paclitaxel binding to proteins in patient ascites because some PM patients show accumulation of ascites and because activity and transport of highly protein-bound drugs such as paclitaxel are affected by protein binding. Ascites samples were collected from 26 patients and investigated for their protein contents using LC/MS/MS proteomics analysis and for the concentrations of total proteins and two major paclitaxel-binding proteins (human serum albumin or HSA and α-1-acid glycoprotein or AAG). The association constants of paclitaxel to HSA and AAG and the extent of protein binding of paclitaxel in patient ascites were studied using equilibrium dialysis. Proteomic analysis of four randomly selected samples revealed 288 proteins, >90% of which are also present in human plasma. Between 72% - 94% of paclitaxel was bound to proteins in patient ascites. The concentrations of HSA and AAG in ascites showed substantial inter-subject variations, ranging from 14.7 - 46.3 mg/mL and 0.13-2.56 mg/mL, respectively. The respective paclitaxel association constants to commercially available HSA and AAG were ∼ 3.5 and ∼ 120 mM. Calculation using these constants and the HSA and AAG concentrations in individual patient ascites indicated that these two proteins accounted for >85% of the total protein-binding of paclitaxel in ascites. The extensive drug binding to ascites proteins, by reducing the pharmacologically active free fraction, may lead to the diminished CRS efficacy in PM patients with ascites. Clinical advances in CRS + IPC have outpaced current knowledge of pharmaceutical properties in this setting. IPC, as a locally acting therapy, is subjected to processes different from those governing systemic treatments. This study, to our knowledge, is the first to illustrate the implications of drug properties in the CRS + IPC efficacy against PM. While drugs are now an integral part of PM patient management, there is limited pharmaceutical research in this treatment setting (e.g., effects of hyperthermia or pressure on drug transport or release from delivery systems, pharmacokinetics, pharmacodynamics). Hence, CRS + IPC of PM represents an area where additional pharmaceutical research can assist further development and optimization.
Collapse
Affiliation(s)
| | - Ze Lu
- Institute of Quantitative Systems Pharmacology, Carlsbad, CA 92008, USA
| | - Carlos H F Chan
- Department of Surgery and Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | - Kiran Turaga
- School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jessie L S Au
- Institute of Quantitative Systems Pharmacology, Carlsbad, CA 92008, USA; Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; College of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
11
|
Ngougni Pokem P, Matzneller P, Vervaeke S, Wittebole X, Goeman L, Coessens M, Cottone E, Capron A, Wulkersdorfer B, Wallemacq P, Mouton JW, Muller AE, Zeitlinger M, Laterre PF, Tulkens PM, Van Bambeke F. Binding of temocillin to plasma proteins in vitro and in vivo: the importance of plasma protein levels in different populations and of co-medications. J Antimicrob Chemother 2022; 77:2742-2753. [PMID: 36018077 DOI: 10.1093/jac/dkac286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/28/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Temocillin plasma protein binding (PPB) in healthy individuals is reported to be ∼85% but had not been studied in patients. OBJECTIVES To obtain normative data on temocillin PPB in patients in relation to infection and impact of co-medications widely used in ICU. METHODS Plasma was obtained from healthy individuals (Group #1), non-ICU patients with UTI (Group #2), ICU patients with suspected/confirmed ventriculitis (Group #3) or with sepsis/septic shock (Group #4). Total and unbound temocillin concentrations were measured in spiked samples from temocillin-naive donors (in vitro) or in plasma from temocillin-treated subjects (in vivo). The impact of diluting plasma, using pharmaceutical albumin, or adding drugs potentially competing for PPB was tested in spiked samples. Data were analysed using a modified Hill-Langmuir equation taking ligand depletion into account. RESULTS Temocillin PPB was saturable in all groups, both in vitro and in vivo. Maximal binding capacity (Bmax) was 1.2-2-fold lower in patients. At 20 and 200 mg/L (total concentrations), the unbound fraction reached 12%-29%, 23%-42% and 32%-52% in Groups #2, #3, #4. The unbound fraction was inversely correlated with albumin and C-reactive protein concentrations. Binding to albumin was 2-3-fold lower than in plasma and non-saturable. Drugs with high PPB but active at lower molar concentrations than temocillin caused minimal displacement, while fluconazole (low PPB but similar plasma concentrations to temocillin) increased up to 2-fold its unbound fraction. CONCLUSIONS Temocillin PPB is saturable, 2-4-fold lowered in infected patients in relation to disease severity (ICU admission, hypoalbuminaemia, inflammation) and only partially reproducible with albumin. Competition with other drugs must be considered for therapeutic concentrations to be meaningful.
Collapse
Affiliation(s)
- Perrin Ngougni Pokem
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Peter Matzneller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna Austria
| | - Steven Vervaeke
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Xavier Wittebole
- Department of Critical Care Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Lieven Goeman
- Department of Urology, AZ Delta Hospital, Roeselare, Belgium
| | - Marie Coessens
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Eleonora Cottone
- Department of Laboratory Medicine, AZ Delta Hospital, Roeselare, Belgium
| | - Arnaud Capron
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | | | - Pierre Wallemacq
- Department of Clinical Chemistry, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, Rotterdam, The Netherlands.,Department of Medical Microbiology, Haaglanden Medical Centre, The Hague, The Netherlands
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna Austria
| | - Pierre François Laterre
- Department of Critical Care Medicine, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Paul M Tulkens
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
12
|
Protein Binding in Translational Antimicrobial Development-Focus on Interspecies Differences. Antibiotics (Basel) 2022; 11:antibiotics11070923. [PMID: 35884177 PMCID: PMC9311574 DOI: 10.3390/antibiotics11070923] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/07/2022] Open
Abstract
Background/Introduction: Plasma protein binding (PPB) continues to be a key aspect of antibiotic development and clinical use. PPB is essential to understand several properties of drug candidates, including antimicrobial activity, drug-drug interaction, drug clearance, volume of distribution, and therapeutic index. Focus areas of the review: In this review, we discuss the basics of PPB, including the main drug binding proteins i.e., Albumin and α-1-acid glycoprotein (AAG). Furthermore, we present the effects of PPB on the antimicrobial activity of antibiotics and the current role of PPB in in vitro pharmacodynamic (PD) models of antibiotics. Moreover, the effect of PPB on the PK/PD of antibiotics has been discussed in this review. A key aspect of this paper is a concise evaluation of PPB between animal species (dog, rat, mouse, rabbit and monkey) and humans. Our statistical analysis of the data available in the literature suggests a significant difference between antibiotic binding in humans and that of dogs or mice, with the majority of measurements from the pre-clinical species falling within five-fold of the human plasma value. Conversely, no significant difference in binding was found between humans and rats, rabbits, or monkeys. This information may be helpful for drug researchers to select the most relevant animal species in which the metabolism of a compound can be studied for extrapolating the results to humans. Furthermore, state-of-the-art methods for determining PPB such as equilibrium dialysis, ultracentrifugation, microdialysis, gel filtration, chromatographic methods and fluorescence spectroscopy are highlighted with their advantages and disadvantages.
Collapse
|
13
|
Turner NA, Xu A, Zaharoff S, Holland TL, Lodise TP. Determination of plasma protein binding of dalbavancin. J Antimicrob Chemother 2022; 77:1899-1902. [PMID: 35488862 PMCID: PMC9633717 DOI: 10.1093/jac/dkac131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/22/2022] [Indexed: 09/20/2023] Open
Abstract
OBJECTIVES Dalbavancin is a lipoglycopeptide with a long half-life, making it a promising treatment for infections requiring prolonged therapy, such as complicated Staphylococcus aureus bacteraemia. Free drug concentration is a critical consideration with prolonged treatment, since free concentration-time profiles may best correlate with therapeutic effect. In support of future clinical trials, we aimed to develop a reliable and reproducible assay for measuring free dalbavancin concentrations. METHODS The ultracentrifugation technique was used to determine free dalbavancin concentrations in plasma at two concentrations (50 and 200 mg/L) in duplicate. Centrifuge tubes and pipette tips were treated for 24 h before use with Tween 80 to assess adsorption. Dalbavancin concentrations were analysed from the plasma samples (total) and middle layer samples (free) by LC/MS/MS with isotopically labelled internal standard. Warfarin served as a positive control with known high protein binding. RESULTS Measurement of free dalbavancin was sensitive to adsorption onto plastic. Treatment of tubes and pipette tips with ≥2% Tween 80 effectively prevented drug loss during protein binding experiments. By the ultracentrifugation method, dalbavancin's protein binding was estimated to be approximately 99%. CONCLUSIONS Dalbavancin has very high protein binding. Given dalbavancin's high protein binding, accurate measurement of free dalbavancin concentrations should be a key consideration in future exposure-response studies, especially clinical trials. Future investigations should confirm if the active fraction is best predicted by the free or total fraction.
Collapse
Affiliation(s)
- Nicholas A Turner
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Allan Xu
- Keystone Bioanalytical, North Wales, PA, USA
| | | | - Thomas L Holland
- Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Duke Clinical Research Institute, Durham, NC, USA
| | - Thomas P Lodise
- Albany College of Pharmacy and Health Sciences, Albany, NY, USA
| |
Collapse
|
14
|
Humphreys SC, Davis JA, Iqbal S, Kamel A, Kulmatycki K, Lao Y, Liu X, Rodgers J, Snoeys J, Vigil A, Weng Y, Wiethoff C, Wittwer M. Considerations and recommendations for assessment of plasma protein binding and drug-drug interactions for siRNA therapeutics. Nucleic Acids Res 2022; 50:6020-6037. [PMID: 35687098 PMCID: PMC9226521 DOI: 10.1093/nar/gkac456] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/25/2022] [Accepted: 06/06/2022] [Indexed: 11/12/2022] Open
Abstract
At the time of writing, although siRNA therapeutics are approved for human use, no official regulatory guidance specific to this modality is available. In the absence of guidance, preclinical development for siRNA followed a hybrid of the small molecule and biologics guidance documents. However, siRNA differs significantly from small molecules and protein-based biologics in its physicochemical, absorption, distribution, metabolism and excretion properties, and its mechanism of action. Consequently, certain reports typically included in filing packages for small molecule or biologics may benefit from adaption, or even omission, from an siRNA filing. In this white paper, members of the 'siRNA working group' in the IQ Consortium compile a list of reports included in approved siRNA filing packages and discuss the relevance of two in vitro reports-the plasma protein binding evaluation and the drug-drug interaction risk assessment-to support siRNA regulatory filings. Publicly available siRNA approval packages and the literature were systematically reviewed to examine the role of siRNA plasma protein binding and drug-drug interactions in understanding pharmacokinetic/pharmacodynamic relationships, safety and translation. The findings are summarized into two decision trees to help guide industry decide when in vitro siRNA plasma protein binding and drug-drug interaction studies are warranted.
Collapse
Affiliation(s)
| | - John A Davis
- PKS Department, Novartis, Cambridge, MA 02139, USA
| | | | - Amin Kamel
- Global DMPK, Takeda, San Diego, CA 92121, USA
| | | | - Yanbin Lao
- DMPK, Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, NJ 08648, USA
| | - Xiumin Liu
- Early Development, Alnylam Pharmaceuticals, Inc., Cambridge, MA 02142, USA
| | - John Rodgers
- PKDM Department, Amgen Inc., South San Francisco, CA 94080, USA
| | - Jan Snoeys
- DMPK Department, Janssen R&D, Beerse 2340, Belgium
| | - Adam Vigil
- DMPK, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT 06877, USA
| | - Yan Weng
- Early Clinical Development Clinical Pharmacology Department, Pfizer, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
15
|
Kondampati KD, Saini SPS, Sidhu PK, Anand A, Kumar D, Srinu B, Bedi JS, Kaur R, Bhardwaj R. PHARMACOKINETIC-PHARMACODYNAMIC STUDY OF AMPICILLIN-CLOXACILLIN COMBINATION IN INDIAN THOROUGHBRED HORSES (Equus caballus) AND SAFETY EVALUATION OF THE COMPUTED DOSAGE REGIMEN. J Equine Vet Sci 2022; 115:104020. [DOI: 10.1016/j.jevs.2022.104020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 10/18/2022]
|
16
|
Peters M, Bockfeld D, Tamm M. Cationic Iridium(I) NHC‐Phosphinidene Complexes and Their Application in Hydrogen Isotope Exchange Reactions. Eur J Inorg Chem 2022. [DOI: 10.1002/ejic.202200148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Marius Peters
- Technische Universität Braunschweig: Technische Universitat Braunschweig Institut für Anorganische und Analytische Chemie GERMANY
| | - Dirk Bockfeld
- Technische Universität Braunschweig: Technische Universitat Braunschweig Institut für Anorganische und Analytische Chemie GERMANY
| | - Matthias Tamm
- Technische Universität Braunschweig Institut für Anorganische und Analytische Chemie Hagenring 30 38106 Braunschweig GERMANY
| |
Collapse
|
17
|
Baruah P, Paul D, Doshi J, Mitra S. Elevated Fibrinogen Level Reduces Therapeutic Efficiency of AD Drugs: Biophysical Insights into the Interaction of FDA-Approved Cholinesterase Inhibitors with Human Fibrinogen. J Phys Chem B 2021; 126:30-43. [PMID: 34964643 DOI: 10.1021/acs.jpcb.1c07495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite being the second most abundant protein in blood plasma, reports on the interaction of drugs with fibrinogen (FIB) are relatively scarce. The effect of FIB on the therapeutic potency of four FDA-approved Alzheimer's disease drugs, namely, tacrine (TAC), donepezil (DON), eserine (ESE), and huperzine (HUP), was investigated through a combination of different in vitro and in silico experiments. The efficiency of the drugs in inhibiting the activity of acetylcholinesterase (AChE) was significantly reduced in the presence of FIB. This effect was even found to be more substantial than that for the most abundant plasma protein, human serum albumin (HSA). For example, the relative change in IC50 for TAC was found to be 65% in 10 μM FIB as opposed to 43% in the presence of 250 μM HSA. The relative trend of modulation in AChE activity showed consistency with the binding efficiency of the drugs and FIB. The sequestration of drugs in FIB, therefore reducing the availability of free drugs in solution, was identified to be the primary cause for the decrease in the AChE inhibition potency. This study aims to establish FIB as a vital component, while considering the therapeutic effectiveness of different newly developed AChE inhibitors.
Collapse
Affiliation(s)
- Prayasee Baruah
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, India
| | - Debojit Paul
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Jitesh Doshi
- BioInsight Solutions (OPC) Pvt. Ltd, Navi Mumbai 410210, India
| | - Sivaprasad Mitra
- Department of Chemistry, North-Eastern Hill University, Shillong 793022, India
| |
Collapse
|
18
|
Hu Y, Li H, Wu M, Zhang H, Ding Y, Peng Y, Li X, Yu Z. Single and multiple dose pharmacokinetics and safety of ZSP1273, an RNA polymerase PB2 protein inhibitor of the influenza A virus: a phase 1 double-blind study in healthy subjects. Expert Opin Investig Drugs 2021; 30:1159-1167. [PMID: 34654349 DOI: 10.1080/13543784.2021.1994944] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
BACKGROUND Influenza is an acute respiratory illness. Treating with antiviral drugs can decrease the duration of illness and serious complications . ZSP1273 is a small-molecule anti-influenza drug targeting the RNA polymerase PB2 subunit of the influenza virus. The aim of this clinical trial was to evaluate the safety and pharmacokinetics (PKs) of ZSP1273 in healthy subjects. RESEARCH DESIGN AND METHODS This was a double-blind, placebo-controlled phase 1 study consisting of three parts. 100 volunteers were enrolled and randomized to receive either single or multiple doses of ZSP1273 or placebo. RESULTS A total of 31 (31.0%) subjects experienced at least one mild or moderate adverse event. The linear regression relationship between dose and plasma Cmax, AUC0-t, and AUC0-∞ showed an increasing trend and rapid absorption of ZSP1273. A high-fat diet had little effect on the PKs. The plasma concentration of ZSP1273 reached steady state on day 5 without drug accumulation. CONCLUSIONS ZSP1273 was safe in healthy volunteers. Based on the preclinical resuilts, safety profile and PK characteristics of ZSP1273, the dose of ZSP1273 (≥200 mg) may be used for future clinical trials in influenza patients. TRIAL REGISTRATION The trial is registered at ClinicalTrials.gov (CT.gov identifier: NCT03679143).
Collapse
Affiliation(s)
- Yue Hu
- Department of Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Haijun Li
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
| | - Min Wu
- Department of Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Hong Zhang
- Department of Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Yanhua Ding
- Department of Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Yun Peng
- Guangdong Raynovent Biotech Co., Ltd, Guangzhou, China
| | - Xiaojiao Li
- Department of Phase I Clinical Trial Unit, The First Hospital of Jilin University, Changchun, China
| | - Zhenxiang Yu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Gad MZ, Azab SS, Khattab AR, Farag MA. Over a century since ephedrine discovery: an updated revisit to its pharmacological aspects, functionality and toxicity in comparison to its herbal extracts. Food Funct 2021; 12:9563-9582. [PMID: 34533553 DOI: 10.1039/d1fo02093e] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ephedrine, a sympathomimetic amine that exhibits several adrenaline actions, is a plant alkaloid that is a common ingredient in several cold, asthma and narcolepsy treatment preparations, and in obesity management and sport medicine. Its principal action mechanism relies on its direct adrenergic actions as well as indirect role that involves the release of epinephrine and norepinephrine, thus increasing the activity of epinephrine and norepinephrine at the postsynaptic α and β receptors. Nevertheless, its serious side effects, including stroke, heart attack, drug abuse and interactions, have never been comprehensively reviewed. We conducted a systematic review of data on ephedrine, including its occurrence in functional foods, pharmacological aspects, metabolism, pharmaco/toxicokinetics and clinical features. Furthermore, a review of ephedrine natural structural analogues with regards to their differential adrenergic receptor binding affinities, food interaction, and their impact on the pharmacokinetics and effects relative to ephedrine are presented for the first time, and in comparison to its action when present in herbs.
Collapse
Affiliation(s)
- Mohamed Z Gad
- Department of Biochemistry, Faculty of Pharmacy & Biotechnology, The German University in Cairo, Egypt
| | - Samar S Azab
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Amira R Khattab
- Pharmacognosy Department, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport, Alexandria 1029, Egypt
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Kasr El Aini St, P.B. 11562, Cairo, Egypt. .,Chemistry Department, School of Sciences & Engineering, The American University in Cairo, New Cairo 11835, Egypt
| |
Collapse
|
20
|
Pantaleão SQ, Fernandes PO, Gonçalves JE, Maltarollo VG, Honorio KM. Recent Advances in the Prediction of Pharmacokinetics Properties in Drug Design Studies: A Review. ChemMedChem 2021; 17:e202100542. [PMID: 34655454 DOI: 10.1002/cmdc.202100542] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/07/2021] [Indexed: 12/11/2022]
Abstract
This review presents the main aspects related to pharmacokinetic properties, which are essential for the efficacy and safety of drugs. This topic is very important because the analysis of pharmacokinetic aspects in the initial design stages of drug candidates can increase the chances of success for the entire process. In this scenario, experimental and in silico techniques have been widely used. Due to the difficulties encountered with the use of some experimental tests to determine pharmacokinetic properties, several in silico tools have been developed and have shown promising results. Therefore, in this review, we address the main free tools/servers that have been used in this area, as well as some cases of application. Finally, we present some studies that employ a multidisciplinary approach with synergy between in silico, in vitro, and in vivo techniques to assess ADME properties of bioactive substances, achieving successful results in drug discovery and design.
Collapse
Affiliation(s)
- Simone Q Pantaleão
- Centro de Ciências Naturais e Humanas, Institution Universidade Federal do ABC, 09210-580, Santo André, SP, Brazil
| | - Philipe O Fernandes
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - José Eduardo Gonçalves
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Vinícius G Maltarollo
- Departamento de Produtos Farmacêuticos, Universidade Federal de Minas Gerais, 31270-901, Pampulha, MG, Brazil
| | - Kathia Maria Honorio
- Centro de Ciências Naturais e Humanas, Institution Universidade Federal do ABC, 09210-580, Santo André, SP, Brazil.,Escola de Artes, Ciências e Humanidades, Universidade de São Paulo, 03828-000, São Paulo, SP, Brazil
| |
Collapse
|
21
|
Towards prevention of ischemia-reperfusion kidney injury: Pre-clinical evaluation of 6-chromanol derivatives and the lead compound SUL-138 ✰. Eur J Pharm Sci 2021; 168:106033. [PMID: 34610451 DOI: 10.1016/j.ejps.2021.106033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 09/06/2021] [Accepted: 10/01/2021] [Indexed: 11/21/2022]
Abstract
Acute kidney injury (AKI) is a global healthcare burden attributable to high mortality and staggering costs of dialysis. The underlying causes of AKI include hypothermia and rewarming (H/R), ischemia/reperfusion (I/R), mitochondrial dysfunction and reactive oxygen species production. Inspired by the mechanisms conferring organ protection in hibernating hamster, 6-chromanol derived compounds were developed to address the need of effective prevention and treatment of AKI. Here we report on the pre-clinical screening of 6-chromanol leads that confer protection during I/R to select compounds with favorable profiles for clinical testing in AKI. A library of 6-chromanols (n = 63) was screened in silico for pharmacochemical properties and druggability. Selected compounds (n = 15) were screened for the potency to protect HEK293 cells from H/R cell death and subjected to a panel of in vitro safety assays. Based on these parameters, SUL-138 was selected as the lead compound and was found to safeguard kidney function and decrease renal injury after I/R in rats. The compound was without cardiovascular or respiratory effects in vivo. SUL-138 pharmacokinetics of control animals (mouse, rat) and those undergoing I/R (rat) was identical, showing a two-phase elimination profile with terminal half-life of about 8 h. Collectively, our phenotype-based screening approach led to the identification of 3 candidates for pre-clinical studies (5%, 3/64). SUL-138 emerged from this small-scale library of 6-chromanols as a novel prophylactic for AKI. The presented efficacy and safety data provide a basis for future development and clinical testing. SECTION ASSIGNMENTS: : Drug discovery and translational medicine, renal, metabolism SIGNIFICANCE STATEMENT: : Based on in silico druggability parameters, a 63 compound 6-chromanol library was narrowed down to 15 compounds. These compounds were subjected to phenotypical screening of cell survival following hypothermia damage and hit compounds were identified. After subsequent assessment of in vivo efficacy, toxicity, pharmacokinetics, and cardiovascular and respiratory safety, SUL-138 emerged as a lead compound that prevented kidney injury after ischemia/reperfusion and demonstrated a favorable pharmacokinetic profile unaffected by renal ischemia.
Collapse
|
22
|
Vay M, Meyer MJ, Blank A, Skopp G, Rose P, Tzvetkov MV, Mikus G. Oral Yohimbine as a New Probe Drug to Predict CYP2D6 Activity: Results of a Fixed-Sequence Phase I Trial. Clin Pharmacokinet 2021; 59:927-939. [PMID: 32060866 PMCID: PMC7329762 DOI: 10.1007/s40262-020-00862-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective Yohimbine pharmacokinetics were determined after oral administration of a single oral dose of yohimbine 5 mg and a microdose of yohimbine 50 µg in relation to different cytochrome P450 (CYP) 2D6 genotypes. The CYP2D6 inhibitor paroxetine was used to investigate the influence on yohimbine pharmacokinetics. Microdosed midazolam was applied to evaluate a possible impact of yohimbine on CYP3A activity and the possibility of combining microdosed yohimbine and midazolam to simultaneously determine CYP2D6 and CYP3A activity. Methods In a fixed-sequence clinical trial, 16 healthy volunteers with a known CYP2D6 genotype [extensive (10), intermediate (2) and poor (4) metaboliser] received an oral dose of yohimbine 50 µg, yohimbine 5 mg at baseline and during paroxetine as a CYP2D6 inhibitor. Midazolam (30 µg) was co-administered to determine CYP3A activity at each occasion. Plasma concentrations of yohimbine, its main metabolite 11-OH-yohimbine, midazolam and paroxetine were quantified using validated liquid chromatography-tandem mass spectrometry assays. Results Pharmacokinetics of yohimbine were highly variable and a CYP2D6 genotype dependent clearance was observed. After yohimbine 5 mg, the clearance ranged from 25.3 to 15,864 mL/min and after yohimbine 50 µg, the clearance ranged from 39.6 to 38,822 mL/min. A more than fivefold reduction in clearance was caused by paroxetine in CYP2D6 extensive metabolisers, while the clearance in poor metabolisers was not affected. Yohimbine did not alter CYP3A activity as measured by microdosed midazolam. Conclusions The pharmacokinetics of yohimbine were highly correlated with CYP2D6, which was further supported by the clearance inhibition caused by the CYP2D6 inhibitor paroxetine. With these data, yohimbine is proposed to be a suitable probe drug to predict CYP2D6 activity. In addition, the microdose can be used in combination with microdosed midazolam to simultaneously evaluate CYP2D6 and CYP3A activity without any interaction between the probe drugs and because the microdoses exert no pharmacological effects. Clinical Trial Registration EudraCT2017-001801-34. Electronic supplementary material The online version of this article (10.1007/s40262-020-00862-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuela Vay
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Marleen Julia Meyer
- Institute of Clinical Pharmacology, University Medical Center Göttingen, Göttingen, Germany
| | - Antje Blank
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | | | - Peter Rose
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | | | - Gerd Mikus
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
23
|
Roy KS, Nazdrajić E, Shimelis OI, Ross MJ, Chen Y, Cramer H, Pawliszyn J. Optimizing a High-Throughput Solid-Phase Microextraction System to Determine the Plasma Protein Binding of Drugs in Human Plasma. Anal Chem 2021; 93:11061-11065. [PMID: 34353028 DOI: 10.1021/acs.analchem.1c01986] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Plasma protein binding refers to the binding of a drug to plasma proteins after entering the body. The measurement of plasma protein binding is essential during drug development and in clinical practice, as it provides a more detailed understanding of the available free concentration of a drug in the blood, which is in turn critical for pharmacokinetics and pharmacodynamics studies. In addition, the accurate determination of the free concentration of a drug in the blood is also highly important for therapeutic drug monitoring and in personalized medicine. The present study uses C18-coated solid-phase microextraction 96-pin devices to determine the free concentrations of a set of drugs in plasma, as well as the plasma protein binding of drugs with a wide range of physicochemical properties. It should be noted that the extracted amounts used to calculate the binding constants and plasma protein bindings should be measured at respective equilibrium for plasma and phosphate buffer. Therefore, special attention is placed on properly determining the equilibration times required to correctly estimate the free concentrations of drugs in the investigated systems. The plasma protein binding values obtained with the 96-pin devices are consistent with those reported in the literature. The 96-pin device used in this research can be easily coupled with a Concept96 or other automated robotic systems to create an automated plasma protein binding determination protocol that is both more time and labor efficient compared to conventional equilibrium dialysis and ultrafiltration methods.
Collapse
Affiliation(s)
- Kanchan Sinha Roy
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Emir Nazdrajić
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Olga I Shimelis
- MilliporeSigma, 595 N. Harrison Road, Bellefonte, Pennsylvania 16823, United States
| | - M James Ross
- MilliporeSigma, 595 N. Harrison Road, Bellefonte, Pennsylvania 16823, United States
| | - Yong Chen
- MilliporeSigma, 595 N. Harrison Road, Bellefonte, Pennsylvania 16823, United States
| | - Hugh Cramer
- MilliporeSigma, 595 N. Harrison Road, Bellefonte, Pennsylvania 16823, United States
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
24
|
Xu M, Zhang H, Tang T, Zhou J, Zhou W, Tan S, He B. Potential and applications of capillary electrophoresis for analyzing traditional Chinese medicine: a critical review. Analyst 2021; 146:4724-4736. [PMID: 34269779 DOI: 10.1039/d1an00767j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Capillary electrophoresis (CE) presents a promising possibility for analyzing traditional Chinese medicine (TCM) due to its low reagent consumption, high analysis speed, and enhanced efficiency. Herein we review the employment of CE for analyzing the effective components in TCM and identifying TCM via a fingerprint. Furthermore, we discuss the application of state-of-the-art capillary electrophoresis modes for screening enzyme inhibitors and investigating the interactions between TCM and plasma proteins. The review concludes with recommendations for future studies and improvements in this field of research. The general development trend identified in this review indicates that the application of CE has significantly improved TCM assay performance.
Collapse
Affiliation(s)
- Mengchang Xu
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Hanyong Zhang
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Tong Tang
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Ji Zhou
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Wenhu Zhou
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Songwen Tan
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| | - Binsheng He
- Academician Workstation, Changsha Medical University, Changsha 410219, China.
| |
Collapse
|
25
|
Ahmad S, Baker D, Murnane D, Spooner N, Gerhard U. Solid-phase microextraction for assessment of plasma protein binding, a complement to rapid equilibrium dialysis. Bioanalysis 2021; 13:1101-1111. [PMID: 34275331 DOI: 10.4155/bio-2021-0109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Determination of plasma protein binding (PPB) is considered vital for better understanding of pharmacokinetic and pharmacodynamic activities of drugs due to the role of free concentration in pharmacological response. Methodology & results: Solid-phase microextraction (SPME) was investigated for measurement of PPB from biological matrices and compared with a gold standard approach (rapid equilibrium dialysis [RED]). Discussion & conclusion: SPME-derived values of PPB correlated well with literature values, and those determined by RED. Respectively, average protein binding across three concentrations by RED and SPME was 33.1 and 31.7% for metoprolol, 89.0 and 86.6% for propranolol and 99.2 and 99.0% for diclofenac. This study generates some evidence for SPME as an alternative platform for the determination of PPB.
Collapse
Affiliation(s)
- Sheelan Ahmad
- Research & Development, GlaxoSmithKline, Stevenage, UK
- Department of Clinical, School of Life & Medical Sciences, Pharmaceutical & Biological Sciences, University of Hertfordshire, Hatfield, UK
| | - Daniel Baker
- Department of Clinical, School of Life & Medical Sciences, Pharmaceutical & Biological Sciences, University of Hertfordshire, Hatfield, UK
| | - Darragh Murnane
- Department of Clinical, School of Life & Medical Sciences, Pharmaceutical & Biological Sciences, University of Hertfordshire, Hatfield, UK
| | - Neil Spooner
- Department of Clinical, School of Life & Medical Sciences, Pharmaceutical & Biological Sciences, University of Hertfordshire, Hatfield, UK
- Spooner Bioanalytical Solutions Ltd., Hertford, UK
| | - Ute Gerhard
- Department of Clinical, School of Life & Medical Sciences, Pharmaceutical & Biological Sciences, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
26
|
Dash RP, Thomas JA, Rosenfeld C, Srinivas NR. Protein Binding and Stability of Drug Candidates: The Achilles' Heel in In Vitro Potency Assays. Eur J Drug Metab Pharmacokinet 2021; 45:427-432. [PMID: 32270425 DOI: 10.1007/s13318-020-00619-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In the present scenario of drug discovery, several screening filters ensure a rigorous nomination of clinical candidates. One of these screens is the determination of IC50, the concentration of drug at half-maximal inhibitory concentration, also known as a potency assay. However, various nuances pertaining to the design, execution, and interpretation of in vitro potency results suggest a sizeable opportunity for the generation of erroneous data. The focus areas of this article include: (1) examining the requirement for the addition of serum albumin in in vitro potency assays, (2) problems encountered with cell lysates, and (3) drug candidate stability concerns during in vitro potency assays/high-throughput screening. Based on this assessment, the interpretation of the data generated using cell-based systems (i.e., lysates with or without the addition of fetal bovine serum) should be carried out with caution for in vitro potency testing, and the inclusion of a correction factor for non-specific protein binding should be considered. The addition of serum albumin to a cell-free system should be restricted to drugs having high protein binding (≥ 90%). Additionally, stability assessment of analytes should be considered to avoid dubious in vitro potency outcomes due to degraded material or active metabolite(s).
Collapse
Affiliation(s)
- Ranjeet P Dash
- ADME-DMPK, Charles River Laboratories, Ashland, OH, 44805, USA
| | | | | | - Nuggehally R Srinivas
- Department of Innovation and Technology, Jubilant Life Sciences, D-12 Sector 59a, Noida, Uttar Pradesh, 201301, India. .,Kenox Pharmaceuticals Inc., 11 Deerpark Dr, Suite 128, Princeton Corporate Plaza, Monmouth Junction, NJ, 08852, USA.
| |
Collapse
|
27
|
Wanat K, Żydek G, Hekner A, Brzezińska E. In silico Plasma Protein Binding Studies of Selected Group of Drugs Using TLC and HPLC Retention Data. Pharmaceuticals (Basel) 2021; 14:ph14030202. [PMID: 33671019 PMCID: PMC7997166 DOI: 10.3390/ph14030202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 11/21/2022] Open
Abstract
Plasma protein binding is an important determinant of the pharmacokinetic properties of chemical compounds in living organisms. The aim of the present study was to determine the index of protein binding affinity based on chromatographic experiments. The question is which chromatographic environment will best mimic the drug–protein binding conditions. Retention data from normal phase thin-layer liquid chromatography (NP TLC), reversed phase (RP) TLC and HPLC chromatography experiments with 129 active pharmaceutical ingredients (APIs) were collected. The stationary phase of the TLC plates was modified with protein and the HPLC column was filled with immobilized human serum albumin. In both chromatographic methods, the mobile phase was based on a buffer with a pH of 7.4 to mimic physiological conditions. Chemometric analyses were performed to compare multiple linear regression models (MLRs) with retention data, using protein binding values as the dependent variable. In the course of the analysis, APIs were divided into acidic, basic and neutral groups, and separate models were created for each group. The MLR models had a coefficient of determination between 0.73 and 0.91, with the highest values from NP TLC data.
Collapse
Affiliation(s)
- Karolina Wanat
- Correspondence: ; Tel.: +48-608-717-573 or +48-42-677-92-11
| | | | | | | |
Collapse
|
28
|
Izat N, Sahin S. Hepatic transporter-mediated pharmacokinetic drug-drug interactions: Recent studies and regulatory recommendations. Biopharm Drug Dispos 2021; 42:45-77. [PMID: 33507532 DOI: 10.1002/bdd.2262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 12/16/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Transporter-mediated drug-drug interactions are one of the major mechanisms in pharmacokinetic-based drug interactions and correspondingly affecting drugs' safety and efficacy. Regulatory bodies underlined the importance of the evaluation of transporter-mediated interactions as a part of the drug development process. The liver is responsible for the elimination of a wide range of endogenous and exogenous compounds via metabolism and biliary excretion. Therefore, hepatic uptake transporters, expressed on the sinusoidal membranes of hepatocytes, and efflux transporters mediating the transport from hepatocytes to the bile are determinant factors for pharmacokinetics of drugs, and hence, drug-drug interactions. In parallel with the growing research interest in this area, regulatory guidances have been updated with detailed assay models and criteria. According to well-established preclinical results, observed or expected hepatic transporter-mediated drug-drug interactions can be taken into account for clinical studies. In this paper, various methods including in vitro, in situ, in vivo, in silico approaches, and combinational concepts and several clinical studies on the assessment of transporter-mediated drug-drug interactions were reviewed. Informative and effective evaluation by preclinical tools together with the integration of pharmacokinetic modeling and simulation can reduce unexpected clinical outcomes and enhance the success rate in drug development.
Collapse
Affiliation(s)
- Nihan Izat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Selma Sahin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
29
|
Yang R, Ren S, Jin X, Sun Y, Dong Y, Zhang J, Liang W, Chen L. Determination of unbound fraction of dorzagliatin in human plasma by equilibrium dialysis and LC-MS/MS and its application to a clinical pharmacokinetic study. J Pharm Biomed Anal 2020; 195:113854. [PMID: 33388639 DOI: 10.1016/j.jpba.2020.113854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/13/2020] [Indexed: 10/22/2022]
Abstract
Dorzagliatin, a novel glucokinase (GK) activator targeting both pancreatic and hepatic GK, is currently in late-stage clinical development for treatment of type 2 diabetes (T2D). For the optimization of dosing regimens to ensure adequate safety and efficacy, it is critical to have a deep understanding of pharmacokinetic (PK) and pharmacodynamic (PD) profiles of the drug in various targeting patient populations, considering the fact that T2D adversely affects a vast patient population who often times also suffer from a wide range of comorbidities including severe liver and/or kidney damage. Since drug efficacy seems to be closely related to unbound drug concentrations at the site of action, therefore, the determination of plasma unbound concentrations/fractions of dorzagliatin is of crucial importance, especially when performing the PK/PD assessment in those special populations. In the current study, a method was developed and validated for determining the unbound fraction (fu) of dorzagliatin in human plasma by using equilibrium dialysis for the separation of the bound and unbound drug, and LC-MS/MS for subsequent quantification. We have successfully addressed two widely recognized challenges for determination of the fu, i.e., the lack of knowledge on the "true fu" and the difficulty in assessing the accuracy and reproducibility of the measurement. Using this method, a 0.2 mL aliquot of human plasma samples were first dialyzed against 0.35 mL of phosphate buffered saline buffer at 37 °C for 5 h in the equilibrium dialysis device to separate the unbound dorzagliatin. Afterwards, post-dialysis samples were extracted by protein precipitation using acetonitrile. Separation of dorzagliatin and potential interferences were achieved using a Gemini C18 column coupled with gradient elution. Subsequent detection was carried out on tandem mass spectrometer operated by multiple reaction monitoring in positive mode using electrospray ionization. The standard curve over the concentration range of 0.125-250 ng/mL exhibits good linearity. The method was fully validated meeting the requirements in current bioanalytical guidance and was successfully applied in a clinical PK study of dorzagliatin in healthy volunteers and patients with renal function impairment. Method reproducibility was demonstrated in incurred sample reanalysis. With demonstrated accuracy, stability and reproducibility, reliable analytical results were obtained from clinical samples for PK/PD interpretation, providing valuable insight for the development of dorzagliatin.
Collapse
Affiliation(s)
- Rong Yang
- Hua Medicine (Shanghai) Ltd., Shanghai, China.
| | - Shuang Ren
- Hua Medicine (Shanghai) Ltd., Shanghai, China
| | - Xiaowei Jin
- Hua Medicine (Shanghai) Ltd., Shanghai, China
| | - Yu Sun
- Hua Medicine (Shanghai) Ltd., Shanghai, China
| | - Yanli Dong
- Hua Medicine (Shanghai) Ltd., Shanghai, China
| | | | | | - Li Chen
- Hua Medicine (Shanghai) Ltd., Shanghai, China.
| |
Collapse
|
30
|
Enabling direct and definitive free fraction determination for highly-bound compounds in protein binding assay. J Pharm Biomed Anal 2020; 194:113765. [PMID: 33272788 DOI: 10.1016/j.jpba.2020.113765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 11/23/2022]
Abstract
Protein binding determination for highly-bound compounds using equilibrium dialysis remains a challenge in drug discovery. The reasons are mainly three-fold; 1. due to their slow diffusion rate, highly-bound compounds require a much longer incubation time to reach dialysis equilibrium than typically needed; 2. highly-bound compounds are often hydrophobic and prone to non-specific binding in dialysis; 3. free drug concentration in the post incubation dialysate is too low for reliable analytical quantification. Modified equilibrium dialysis approaches include using diluted plasma for dialysis, or pre-saturating the non-specific binding sites in the dialysis device with compounds of interest prior to dialysis. In this study, we developed a customized equilibrium dialysis assay with an extended incubation time of 24 h, followed by microflow (μF) LC-MS/MS for bioanalysis, for direct and definitive free fraction determination of highly protein-bound compounds. The extended incubation time ensured the dialysis to reach equilibrium and saturating the non-specific binding sites, while μFLC-MS/MS provided far better sensitivity than the conventional LC-MS/MS typically used for post incubation bioanalysis. For a group of commercially available, highly protein-bound compounds, the free fraction data generated by the developed assay correlated very well with the literature values generated with diluted plasma method or pre-saturation method. This novel assay approach has been successfully used to generate protein binding results for highly-bound compounds to support ongoing drug discovery research.
Collapse
|
31
|
King K, Li P, Pietrasiewicz A, Goldstein S. Perspectives on updates, clarifications and controversies in chromatographic assay guidance for bioanalytical method validation from major regulatory agencies and organizations. Biomed Chromatogr 2020; 35:e5030. [PMID: 33201529 DOI: 10.1002/bmc.5030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 11/10/2022]
Abstract
Bioanalysis, a key supporting function for generating data for pre-clinical and clinical studies in drug development, is under the regulation of local agencies as well as global organizations to ensure the data integrity and quality in submission. As major regulatory agencies and organizations, the US Food and Drug Administration, the European Medicines Agency and the International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use have been updating their industry guidance for bioanalytical method validation, to keep up with the development new modalities, technologies and regulations. This article summarizes the recent updates and any clarifications and controversies triggered by those updates. Perspectives and recommendations are given based on our own experience as well as commonly accepted practice in the bioanalytical community.
Collapse
Affiliation(s)
- Kris King
- Department of Drug Metabolism and Pharmacokinetics, Biogen, 225 Binney St, Cambridge, MA, 02142, USA
| | - Pei Li
- Department of Drug Metabolism and Pharmacokinetics, Biogen, 225 Binney St, Cambridge, MA, 02142, USA
| | - Alicia Pietrasiewicz
- Department of Drug Metabolism and Pharmacokinetics, Biogen, 225 Binney St, Cambridge, MA, 02142, USA
| | - Stanley Goldstein
- Department of Drug Metabolism and Pharmacokinetics, Biogen, 225 Binney St, Cambridge, MA, 02142, USA
| |
Collapse
|
32
|
Bioanalytical aspects and challenges in supporting pediatric drug development. Bioanalysis 2020; 12:1505-1508. [PMID: 33078957 DOI: 10.4155/bio-2020-0237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
33
|
Blood distribution and plasma protein binding of PHOTOCYANINE: a promising phthalocyanine photosensitizer inphaseⅡ clinical trials. Eur J Pharm Sci 2020; 153:105491. [PMID: 32726646 DOI: 10.1016/j.ejps.2020.105491] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/25/2020] [Accepted: 07/25/2020] [Indexed: 12/30/2022]
Abstract
Blood distribution and plasma protein binding are the important properties that can influence pharmacokinetics and ultimately the anticancer efficacy of photosensitizers in clinical photodynamic therapy. As a novel and promising phthalocyanine photosensitizer under clinical phase Ⅱ investigation in China, the superiority of PHOCYANINE is speculated on its attribution to its binding with plasma proteins. To verify this hypothesis, explore the targeting mechanism and further apply foundation for its clinical trial evaluation, we further study its in vitro and in vivo human blood distribution, in vitro plasma protein and lipoprotein binding in detail. PHOTOCYANINE was found to be mainly distributed in plasma with low KBP and KEP values. Moreover, its high binding rates to plasma proteins among various species (mouse, rat, dog, monkey, and human) were then determined. Among these plasma proteins, human serum albumin and α1-acid-glycoprotein were found to bind PHOTOCYANINE highly, and low-density lipoproteins have the highest percentage of PHOTOCYANINE over other lipoproteins. This study is expected to provide some guidance for PDT clinical evaluations and for further molecular design and development of photosensitizers.
Collapse
|
34
|
Legros F, Fernandez‐Rodriguez P, Mishra A, Weck R, Bauer A, Sandvoss M, Ruf S, Méndez M, Mora‐Radó H, Rackelmann N, Pöverlein C, Derdau V. Photoredox‐Mediated Hydrogen Isotope Exchange Reactions of Amino‐Acids, Peptides, and Peptide‐Derived Drugs. Chemistry 2020; 26:12738-12742. [DOI: 10.1002/chem.202003464] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Fabien Legros
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | | | - Anurag Mishra
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Remo Weck
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Armin Bauer
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Martin Sandvoss
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Sven Ruf
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - María Méndez
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Helena Mora‐Radó
- TIDES Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Nils Rackelmann
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Christoph Pöverlein
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| | - Volker Derdau
- Integrated Drug Discovery Sanofi-Aventis (Deutschland) GmbH Industriepark Höchst 65926 Frankfurt Germany
| |
Collapse
|
35
|
Xu L, Lee W, Rotenberg A, Böhlke M, Yoon K, Yoo SS. Localized Disruption of Blood Albumin-Phenytoin Binding Using Transcranial Focused Ultrasound. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1986-1997. [PMID: 32402673 DOI: 10.1016/j.ultrasmedbio.2020.04.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 04/02/2020] [Accepted: 04/10/2020] [Indexed: 06/11/2023]
Abstract
Plasma protein binding (PPB) plays an important role in drug pharmacokinetics, particularly for central nervous system drugs, as PPB affects the blood concentration of unbound drug available to cross the blood-brain barrier (BBB). We report the non-invasive, spatially specific disruption of PPB to phenytoin, an anti-epileptic drug with high affinity to albumin, using 250-kHz focused ultrasound (FUS) delivered in a pulsed manner (55-ms tone burst duration, 4-Hz pulse repetitions). Equilibrium dialysis performed on sonicated phosphate-buffered saline solution containing phenytoin and bovine serum albumin revealed a 27.7% elevation in the unbound phenytoin concentration compared with an unsonicated control. Sonication of a unilateral brain hemisphere in rats (n = 10) after intraperitoneal phenytoin injection revealed increased parenchymal phenytoin uptake compared with the unsonicated hemisphere, without evidence of temperature change or BBB disruption. These findings illustrate the use of FUS as a novel technique for spatially selective disruption of PPB, which may be applied to a wide range of drug-plasma protein interactions.
Collapse
Affiliation(s)
- Linda Xu
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wonhye Lee
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alexander Rotenberg
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark Böhlke
- Massachusetts College of Pharmacy and Health Sciences University, Boston, Massachusetts, USA
| | - Kyungho Yoon
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
36
|
Moein MM, Halldin C. Sample preparation techniques for protein binding measurement in radiopharmaceutical approaches: A short review. Talanta 2020; 219:121220. [PMID: 32887121 DOI: 10.1016/j.talanta.2020.121220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 10/24/2022]
Abstract
Plasma protein binding (PPB) measurement is a key step in radiopharmaceutical studies for the development of positron emission tomography (PET) radioligands. PPB refers to the binding degree of a radioligand, radiotracer, or drug to blood plasma proteins or tissues after administration into the body. Several techniques have been successfully developed and applied for PPB measurement of PET radioligands. However, there is room for progress among these techniques in relation to duration time, adaptability with nonpolar radioligands, in vivo measurement, specificity, and selectivity. This mini review gives a brief overview of advances, limitations, and prospective applications of commercially-available PPB methods.
Collapse
Affiliation(s)
- Mohammad Mahdi Moein
- Karolinska Radiopharmacy, Karolinska University Hospital, S-171 64 Stockholm, Sweden; Karolinska Institutet, Department of Oncology-Pathology, J5:20, S-171 77 Stockholm, Sweden.
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, SE-171 76 Stockholm, Sweden
| |
Collapse
|
37
|
Dogra A, Kotwal P, Gour A, Bhatt S, Singh G, Mukherjee D, Nandi U. Description of Druglike Properties of Safranal and Its Chemistry behind Low Oral Exposure. ACS OMEGA 2020; 5:9885-9891. [PMID: 32391475 PMCID: PMC7203973 DOI: 10.1021/acsomega.0c00160] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/01/2020] [Indexed: 05/11/2023]
Abstract
Safranal, a plant secondary metabolite isolated from saffron, has been reported for several promising pharmacological properties toward the management of Alzheimer's disease. In the present study, we observe and report for the first time about several druglike attributes of safranal, such as adherence to Lipinski's rule of five; optimum lipophilicity; high permeability; low blood-to-plasma ratio; less to moderate propensity to interact with P-glycoprotein (P-gp) or breast cancer-resistant protein (BCRP) transporters; and high plasma protein binding as common to most of the marketed drugs using in vitro and ex vivo models. In spite of the above attributes, in vivo oral absorption was found to be very poor, which is linked to the structural integrity of safranal in simulated gastric fluid, simulated intestinal fluid, plasma, and liver microsomes. Moreover, the presence of unsaturated aldehyde moiety in safranal remains in equilibrium with its hydroxylated acetal form. Further research work is required to find out the stable oral absorbable form of safranal by derivatization of its aldehyde group without losing its potency.
Collapse
Affiliation(s)
- Ashish Dogra
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Pankul Kotwal
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Abhishek Gour
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Shipra Bhatt
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Gurdarshan Singh
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Debaraj Mukherjee
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Natural
Product Chemistry Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
| | - Utpal Nandi
- PK-PD,
Toxicology and Formulation Division, CSIR-Indian
Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine, Jammu, Jammu and Kashmir 180001, India
- ,
| |
Collapse
|
38
|
Rong R, Wang X, Dan Y, Zhang R, Zhao Y, Yu Z. Determination of protein binding for novel 2‐(2‐hydroxypropanamido)‐5‐trifluoromethyl benzoic acid enantiomers to rats, dogs, and humans plasma by UPLC‐MS/MS. Chirality 2020; 32:524-534. [DOI: 10.1002/chir.23192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 11/09/2022]
Affiliation(s)
- Rong Rong
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| | - Xin Wang
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| | - Yuhan Dan
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| | - Ruizhen Zhang
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| | - Yunli Zhao
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| | - Zhiguo Yu
- School of PharmacyShenyang Pharmaceutical University Shenyang Liaoning Province China
| |
Collapse
|
39
|
Lamy E, Fall F, Boigne L, Gromov K, Fabresse N, Grassin-Delyle S. Validation according to European and American regulatory agencies guidelines of an LC-MS/MS method for the quantification of free and total ropivacaine in human plasma. ACTA ACUST UNITED AC 2020; 58:701-708. [DOI: 10.1515/cclm-2018-1298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/05/2019] [Indexed: 11/15/2022]
Abstract
AbstractBackgroundRopivacaine is a widely used local anaesthetic drug, highly bound to plasma proteins with a free plasma fraction of about 5%. Therefore, the monitoring of free drug concentration is most relevant to perform pharmacokinetic studies and to understand the drug pharmacokinetic/pharmacodynamic (PK/PD) relationship.MethodsA high-sensitivity liquid chromatography-tandem mass spectrometry (LC-MS/MS) method using reverse-phase LC and electrospray ionisation mass spectrometry with multiple reaction monitoring (MRM) is described for the quantitation of both free and total ropivacaine in human plasma. Ropivacaine-d7 was used as an internal standard (IS).ResultsThe method was validated in the range 0.5–3000 ng/mL, with five levels of QC samples and according to the European Medicine Agency and Food and Drug Administration guidelines. The performance of the method was excellent with a precision in the range 6.2%–14.7%, an accuracy between 93.6% and 113.7% and a coefficient of variation (CV) of the IS-normalised matrix factor below 15%. This suitability of the method for the quantification of free and total ropivacaine in clinical samples was demonstrated with the analysis of samples from patients undergoing knee arthroplasty and receiving a local ropivacaine infiltration.ConclusionsA method was developed and validated for the quantification of free and total ropivacaine in human plasma and was shown suitable for the analysis of clinical samples.
Collapse
Affiliation(s)
- Elodie Lamy
- Plateforme de spectrométrie de masse MasSpecLab, INSERM UMR 1173, UFR Simone Veil – Santé, Université Versailles Saint Quentin, Université Paris Saclay, Montigny le Bretonneux, France
| | - Fanta Fall
- Plateforme de spectrométrie de masse MasSpecLab, INSERM UMR 1173, UFR Simone Veil – Santé, Université Versailles Saint Quentin, Université Paris Saclay, Montigny le Bretonneux, France
| | - Lisa Boigne
- Plateforme de spectrométrie de masse MasSpecLab, INSERM UMR 1173, UFR Simone Veil – Santé, Université Versailles Saint Quentin, Université Paris Saclay, Montigny le Bretonneux, France
| | - Kirill Gromov
- Department of Orthopaedic Surgery, Copenhagen University Hospital, Hvidovre, Denmark
| | - Nicolas Fabresse
- Plateforme de spectrométrie de masse MasSpecLab, INSERM UMR 1173, UFR Simone Veil – Santé, Université Versailles Saint Quentin, Université Paris Saclay, Montigny le Bretonneux, France
- Laboratoire de Toxicologie, Hôpital Raymond Poincaré, AP-HP, Garches, France
| | - Stanislas Grassin-Delyle
- Plateforme de spectrométrie de masse MasSpecLab, INSERM UMR 1173, UFR Simone Veil – Santé, Université Versailles Saint Quentin, Université Paris Saclay, Montigny le Bretonneux, France
- Département des maladies respiratoires, Hôpital Foch, Suresnes, France, Phone: +33.1.70.42.94.22
| |
Collapse
|
40
|
Fundamental aspects of DMPK optimization of targeted protein degraders. Drug Discov Today 2020; 25:969-982. [PMID: 32298797 DOI: 10.1016/j.drudis.2020.03.012] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/03/2020] [Accepted: 03/16/2020] [Indexed: 12/30/2022]
Abstract
Targeted protein degraders are an emerging modality. Their properties fall outside the traditional small-molecule property space and are in the 'beyond rule of 5' space. Consequently, drug discovery programs focused on developing orally bioavailable degraders are expected to face complex drug metabolism and pharmacokinetics (DMPK) challenges compared with traditional small molecules. Nevertheless, little information is available on the DMPK optimization of oral degraders. Therefore, in this review, we discuss our experience of these DMPK optimization challenges and present methodologies and strategies to overcome the hurdles dealing with this new small-molecule modality, specifically in developing oral degraders to treat cancer.
Collapse
|
41
|
Valero M, Kruissink T, Blass J, Weck R, Güssregen S, Plowright AT, Derdau V. C-H Functionalization-Prediction of Selectivity in Iridium(I)-Catalyzed Hydrogen Isotope Exchange Competition Reactions. Angew Chem Int Ed Engl 2020; 59:5626-5631. [PMID: 31917506 PMCID: PMC7232431 DOI: 10.1002/anie.201914220] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 12/22/2022]
Abstract
An assessment of the C-H activation catalyst [(COD)Ir(IMes)(PPh3 )]PF6 (COD=1,5-cyclooctadiene, IMes=1,3-bis(2,4,6-trimethylphenyl)imidazol-2-ylidene) in the deuteration of phenyl rings containing different functional directing groups is divulged. Competition experiments have revealed a clear order of the directing groups in the hydrogen isotope exchange (HIE) with an iridium (I) catalyst. Through DFT calculations the iridium-substrate coordination complex has been identified to be the main trigger for reactivity and selectivity in the competition situation with two or more directing groups. We postulate that the competition concept found in this HIE reaction can be used to explain regioselectivities in other transition-metal-catalyzed functionalization reactions of complex drug-type molecules as long as a C-H activation mechanism is involved.
Collapse
Affiliation(s)
- Mégane Valero
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Thomas Kruissink
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Jennifer Blass
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Remo Weck
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Stefan Güssregen
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Alleyn T. Plowright
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Volker Derdau
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| |
Collapse
|
42
|
Valero M, Bouzouita D, Palazzolo A, Atzrodt J, Dugave C, Tricard S, Feuillastre S, Pieters G, Chaudret B, Derdau V. NHC-Stabilized Iridium Nanoparticles as Catalysts in Hydrogen Isotope Exchange Reactions of Anilines. Angew Chem Int Ed Engl 2020; 59:3517-3522. [PMID: 31849160 PMCID: PMC7079112 DOI: 10.1002/anie.201914369] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/21/2022]
Abstract
The preparation of N-heterocyclic carbene-stabilized iridium nanoparticles and their application in hydrogen isotope exchange reactions is reported. These air-stable and easy-to-handle iridium nanoparticles showed a unique catalytic activity, allowing selective and efficient hydrogen isotope incorporation on anilines using D2 or T2 as isotopic source. The usefulness of this transformation has been demonstrated by the deuterium and tritium labeling of diverse complex pharmaceuticals.
Collapse
Affiliation(s)
- Mégane Valero
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Donia Bouzouita
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées135, Avenue de Rangueil31077ToulouseFrance
| | - Alberto Palazzolo
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay91191Gif-sur-YvetteFrance
| | - Jens Atzrodt
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| | - Christophe Dugave
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay91191Gif-sur-YvetteFrance
| | - Simon Tricard
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées135, Avenue de Rangueil31077ToulouseFrance
| | | | - Grégory Pieters
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay91191Gif-sur-YvetteFrance
| | - Bruno Chaudret
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées135, Avenue de Rangueil31077ToulouseFrance
| | - Volker Derdau
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug DiscoveryIndustriepark Höchst65926Frankfurt am MainGermany
| |
Collapse
|
43
|
Dargó G, Bajusz D, Simon K, Müller J, Balogh GT. Human Serum Albumin Binding in a Vial: A Novel UV-pH Titration Method To Assist Drug Design. J Med Chem 2020; 63:1763-1774. [PMID: 31995375 PMCID: PMC7307925 DOI: 10.1021/acs.jmedchem.0c00046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
![]()
The knowledge on human serum albumin
(HSA) binding is of utmost
importance as it affects pharmacokinetic behavior and bioavailability
of drugs. In this article, we report a novel method to screen for
ionizable molecules with high HSA binding affinity based on pKa shifts using UV-pH titration. We investigated
the HSA binding of 27 drugs and compared the results to experimental
data from conventional methods. In most cases, significant shifts
(ΔpKa > 0.1) were observed for
drugs
with high HSA binding, while no change could be detected for low-affinity
binders. We showed the pivotal role of ionization centers in the formation
of strong interactions between drug and HSA using molecular docking
studies. We also verified our findings by testing five modified analogues
designed by structural considerations. Significant decreases in their
HSA binding proved that the UV-pH titration method combined with an
in silico support can be used as a medicinal chemistry tool to assist
rational molecular design.
Collapse
Affiliation(s)
- Gergő Dargó
- Department of Chemical and Environmental Process Engineering , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary.,Chemistry Department , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group , Research Centre for Natural Sciences , Magyar tudósok krt. 2 , 1117 Budapest , Hungary
| | - Kristóf Simon
- Department of Organic Chemistry and Technology , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary
| | - Judit Müller
- Medicinal Chemistry Laboratory II , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary
| | - György T Balogh
- Department of Chemical and Environmental Process Engineering , Budapest University of Technology and Economics , Műegyetem rakpart 3 , 1111 Budapest , Hungary.,Chemistry Department , Gedeon Richter Plc. , Gyömrői út. 19-21 , 1107 Budapest , Hungary.,Department of Pharmacodynamics and Biopharmacy , University of Szeged , 6720 Szeged , Hungary
| |
Collapse
|
44
|
Valero M, Kruissink T, Blass J, Weck R, Güssregen S, Plowright AT, Derdau V. C−H Functionalization—Prediction of Selectivity in Iridium(I)‐Catalyzed Hydrogen Isotope Exchange Competition Reactions. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Mégane Valero
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Thomas Kruissink
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Jennifer Blass
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Remo Weck
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Stefan Güssregen
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Alleyn T. Plowright
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Volker Derdau
- Sanofi-Aventis (Deutschland) GmbH, R&D Integrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| |
Collapse
|
45
|
Valero M, Bouzouita D, Palazzolo A, Atzrodt J, Dugave C, Tricard S, Feuillastre S, Pieters G, Chaudret B, Derdau V. NHC‐Stabilized Iridium Nanoparticles as Catalysts in Hydrogen Isotope Exchange Reactions of Anilines. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201914369] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Mégane Valero
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Donia Bouzouita
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées 135, Avenue de Rangueil 31077 Toulouse France
| | - Alberto Palazzolo
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay 91191 Gif-sur-Yvette France
| | - Jens Atzrodt
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| | - Christophe Dugave
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay 91191 Gif-sur-Yvette France
| | - Simon Tricard
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées 135, Avenue de Rangueil 31077 Toulouse France
| | - Sophie Feuillastre
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay 91191 Gif-sur-Yvette France
| | - Grégory Pieters
- SCBMJOLIOT InstituteCEAUniversité Paris-Saclay 91191 Gif-sur-Yvette France
| | - Bruno Chaudret
- LPCNO, Laboratoire de Physique et Chimie de Nano-ObjetsUMR5215 INSA-CNRS-UPSInstitut National des Sciences Appliquées 135, Avenue de Rangueil 31077 Toulouse France
| | - Volker Derdau
- Sanofi-Aventis (Deutschland) GmbH, R&DIntegrated Drug Discovery Industriepark Höchst 65926 Frankfurt am Main Germany
| |
Collapse
|
46
|
Müller V, Weck R, Derdau V, Ackermann L. Ruthenium(II)‐Catalyzed Hydrogen Isotope Exchange of Pharmaceutical Drugs by C−H Deuteration and C−H Tritiation. ChemCatChem 2019. [DOI: 10.1002/cctc.201902051] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Valentin Müller
- Institut für Organische und Biomolekulare ChemieGeorg-August-Universität Göttingen Tammannstrasse 2 Göttingen 37077 Germany
| | - Remo Weck
- R&DIntegrated Drug DiscoveryIsotope ChemistrySanofi-Aventis Deutschland GmbH Industriepark Höchst Frankfurt am Main 65926 Germany
| | - Volker Derdau
- R&DIntegrated Drug DiscoveryIsotope ChemistrySanofi-Aventis Deutschland GmbH Industriepark Höchst Frankfurt am Main 65926 Germany
| | - Lutz Ackermann
- Institut für Organische und Biomolekulare ChemieGeorg-August-Universität Göttingen Tammannstrasse 2 Göttingen 37077 Germany
| |
Collapse
|
47
|
Evaluation of electrophoretic mobility shift assay as a method to determine plasma protein binding of siRNA. Bioanalysis 2019; 11:1927-1939. [DOI: 10.4155/bio-2019-0151] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: The electrophoretic mobility shift assay (EMSA) was evaluated as an alternative to ultrafiltration (UF) to assess plasma protein binding (PPB) of small interfering RNAs (siRNA) and antisense oligonucleotides (ASO). Results & methodology: EMSA analysis showed that PPB depended on siRNA and plasma concentration. Conversely, when analyzed by ultrafiltration, siRNA bound the filtration device nonspecifically and PPB remained >98% across physiologically relevant siRNA concentrations. Using EMSA, siRNA exhibited charge-based interactions with plasma proteins, while ASO remained highly bound to plasma proteins or albumin in the presence of 500 mM salt. Conclusion: PPB characteristics of siRNA and ASO can be distinguished using EMSA. Characterization of siRNA PPB by EMSA enhances our knowledge of siRNA absorption, distribution, metabolism and excretion and advanced development of RNA interference therapeutics.
Collapse
|
48
|
Gao L, Xue Y, Zhang Z, Tian Y. Enantioseparation of
N‐
acetyl‐glutamine enantiomers by LC–MS/MS and its application to a plasma protein binding study. Biomed Chromatogr 2019; 33:e4559. [DOI: 10.1002/bmc.4559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/12/2019] [Accepted: 04/17/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Lei Gao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)China Pharmaceutical University Nanjing China
- Key Laboratory of Drug Consistency EvaluationChina Pharmaceutical University Nanjing China
| | - Yunwen Xue
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)China Pharmaceutical University Nanjing China
- Key Laboratory of Drug Consistency EvaluationChina Pharmaceutical University Nanjing China
| | - Zunjian Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)China Pharmaceutical University Nanjing China
- Key Laboratory of Drug Consistency EvaluationChina Pharmaceutical University Nanjing China
| | - Yuan Tian
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education)China Pharmaceutical University Nanjing China
- Key Laboratory of Drug Consistency EvaluationChina Pharmaceutical University Nanjing China
| |
Collapse
|
49
|
Valero M, Mishra A, Blass J, Weck R, Derdau V. Comparison of Iridium(I) Catalysts in Temperature Mediated Hydrogen Isotope Exchange Reactions. ChemistryOpen 2019; 8:1183-1189. [PMID: 31497473 PMCID: PMC6718078 DOI: 10.1002/open.201900204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Indexed: 11/19/2022] Open
Abstract
The reactivity and selectivity of iridium(I) catalysed hydrogen isotope exchange (HIE) reactions can be varied by using wide range of reaction temperatures. Herein, we have done a detailed comparison study with common iridium(I) catalysts (1–6) which will help us to understand and optimize the approaches of either high selectivity or maximum deuterium incorporation. We have demonstrated that the temperature window for these studied iridium(I) catalysts is surprisingly very broad. This principle was further proven in some HIE reactions on complex drug molecules.
Collapse
Affiliation(s)
- Mégane Valero
- Sanofi-Aventis Deutschland GmbH, Integrated Drug Discovery, Isotope Chemistry Industriepark Höchst Frankfurt Germany
| | - Anurag Mishra
- Sanofi-Aventis Deutschland GmbH, Integrated Drug Discovery, Isotope Chemistry Industriepark Höchst Frankfurt Germany
| | - Jennifer Blass
- Sanofi-Aventis Deutschland GmbH, Integrated Drug Discovery, Isotope Chemistry Industriepark Höchst Frankfurt Germany
| | - Remo Weck
- Sanofi-Aventis Deutschland GmbH, Integrated Drug Discovery, Isotope Chemistry Industriepark Höchst Frankfurt Germany
| | - Volker Derdau
- Sanofi-Aventis Deutschland GmbH, Integrated Drug Discovery, Isotope Chemistry Industriepark Höchst Frankfurt Germany
| |
Collapse
|
50
|
Chen Z, Jiang L. The clinical application of fruquintinib on colorectal cancer. Expert Rev Clin Pharmacol 2019; 12:713-721. [PMID: 31177854 DOI: 10.1080/17512433.2019.1630272] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Zhongguang Chen
- Department of Pharmaceutical, Central Hospital of Linyi City, Yishui, Shandong, China
| | - Lili Jiang
- Ultrasound Medical Department, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|