1
|
Widjaja F, van Boekel MAJS, Davis C, Wesseling S, Rietjens IMCM. Quantifying the effect of human interindividual kinetic differences on the relative potency value for riddelliine N-oxide at low dose levels by a new approach methodology. Regul Toxicol Pharmacol 2024:105767. [PMID: 39710333 DOI: 10.1016/j.yrtph.2024.105767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/15/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Pyrrolizidine alkaloids N-oxides (PA-N-oxides) are predominant in plants and herbal foods, and are converted to pyrrolizidine alkaloids (PAs) upon consumption, leading to toxicity. The effect of interindividual kinetic differences on the relative potency values of PA-N-oxides compared to their PAs (REPPANO to PA) was studied, with riddelliine N-oxide (RIDO) and riddelliine (RID) as model compounds. In vitro kinetic data measured in incubations with 30 fecal and 25 liver S9 donor samples showed high variation across individuals, where the interindividual variability was captured with Bayesian multilevel regression. The distributions of influential PBK model parameters were used as input for physiologically based kinetic (PBK) modeling combined with Monte Carlo (MC) simulations to calculate the probability distribution of REPRIDO to RID values. At low internal dose levels, interindividual differences were shown to be a factor that influences the REPRIDO to RID value while neither dose nor endpoint used plays a role. The distribution of the REPRIDO to RID value ranged from 0.71 to 0.97 (95th percentile) with a mean value of 0.87. The approach described enables determination of interindividual REPPANO to PA values at low dose levels, which are not accessible in in vivo experiments quantifying the REP value.
Collapse
Affiliation(s)
- F Widjaja
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands.
| | - M A J S van Boekel
- Food Quality and Design, Wageningen University, PO Box 8129, 6700 EV Wageningen, the Netherlands
| | - C Davis
- Daiichi Sankyo, Inc. 211 Mt. Airy Rd. Basking Ridge, NJ, 07920, USA
| | - S Wesseling
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| | - I M C M Rietjens
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| |
Collapse
|
2
|
Alhejji Y, Widjaja F, Tian S, Hoekstra T, Wesseling S, Rietjens IM. In vitro-in silico study on the influence of dose, fraction bioactivated and endpoint used on the relative potency value of pyrrolizidine alkaloid N-oxides compared to parent pyrrolizidine alkaloids. Curr Res Toxicol 2024; 6:100160. [PMID: 38469320 PMCID: PMC10926302 DOI: 10.1016/j.crtox.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024] Open
Abstract
Pyrrolizidine alkaloids (PAs) and their N-oxides (PA-N-oxides) are phytotoxins found in food, feed and the environment. Yet, limited data exist from which the relative potency of a PA-N-oxide relative to its corresponding PA (REPPANO to PA) can be defined. This study aims to investigate the influence of dose, fraction bioactivated and endpoint on the REPPANO to PA of a series of pyrrolizidine N-oxides using in vitro-in silico data and physiologically based kinetic (PBK) modeling. The first endpoint used to calculate the REPPANO to PA was the ratio of the area under the concentration-time curve of PA resulting from an oral dose of PA-N-oxide divided by that from an equimolar dose of PA (Method 1). The second endpoint was the ratio of the amount of pyrrole-protein adducts formed under these conditions (Method 2). REPPANO to PA values appeared to decrease with increasing dose, with the decrease for Method 2 already starting at lower dose level than for Method 1. At dose levels as low as estimated daily human intakes, REPPANO to PA values amounted to 0.92, 0.81, 0.78, and 0.68 for retrorsine N-oxide, seneciphylline N-oxide, riddelliine N-oxide and senecivernine N-oxide, respectively, and became independent of the dose or fraction bioactivated, because no GSH depletion, saturation of PA clearance or PA-N-oxide reduction occurs. Overall, the results demonstrate the strength of using PBK modeling in defining REPPANO to PA values, thereby substantiating the use of the same approach for other PA-N-oxides for which in vivo data are lacking.
Collapse
Affiliation(s)
- Yasser Alhejji
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah 51452, Saudi Arabia
| | - Frances Widjaja
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| | - Shenghan Tian
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| | - Thomas Hoekstra
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| | - Ivonne M.C.M. Rietjens
- Division of Toxicology, Wageningen University, PO Box 8000, 6700 EA Wageningen, the Netherlands
| |
Collapse
|
3
|
Padilha EC, Yang M, Shah P, Wang AQ, Duan J, Park JK, Zawatsky CN, Malicdan MCV, Kunos G, Iyer MR, Gaucher G, Ravenelle F, Cinar R, Xu X. In vitro and in vivo pharmacokinetic characterization, chiral conversion and PBPK scaling towards human PK simulation of S-MRI-1867, a drug candidate for Hermansky-Pudlak syndrome pulmonary fibrosis. Biomed Pharmacother 2023; 168:115178. [PMID: 37890204 DOI: 10.1016/j.biopha.2023.115178] [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: 03/13/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 10/29/2023] Open
Abstract
Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disorder that affects lysosome-related organelles, often leading to fatal pulmonary fibrosis (PF). The search for a treatment for HPS pulmonary fibrosis (HPSPF) is ongoing. S-MRI-1867, a dual cannabinoid receptor 1 (CB1R)/inducible nitric oxide synthase (iNOS) inhibitor, has shown great promise for the treatment of several fibrotic diseases, including HPSPF. In this study, we investigated the in vitro ADME characteristics of S-MRI-1867, as well as its pharmacokinetic (PK) properties in mice, rats, dogs, and monkeys. S-MRI-1867 showed low aqueous solubility (< 1 µg/mL), high plasma protein binding (>99%), and moderate to high metabolic stability. In its preclinical PK studies, S-MRI-1867 exhibited moderate to low plasma clearance (CLp) and high steady-state volume of distribution (Vdss) across all species. Despite the low solubility and P-gp efflux, S-MRI-1867 showed great permeability and metabolic stability leading to a moderate bioavailability (21-60%) across mouse, rat, dog, and monkey. Since the R form of MRI-1867 is CB1R-inactive, we investigated the potential conversion of S-MRI-1867 to R-MRI-1867 in mice and found that the chiral conversion was negligible. Furthermore, we developed and validated a PBPK model that adequately fits the PK profiles of S-MRI-1867 in mice, rats, dogs, and monkeys using various dosing regimens. We employed this PBPK model to simulate the human PK profiles of S-MRI-1867, enabling us to inform human dose selection and support the advancement of this promising drug candidate in the treatment of HPSPF.
Collapse
Affiliation(s)
- Elias C Padilha
- Drug Metabolism and Pharmacokinetics Core, National Center for Advancing Translational Sciences, Rockville, MD, USA.
| | - Mengbi Yang
- Drug Metabolism and Pharmacokinetics Core, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Pranav Shah
- Drug Metabolism and Pharmacokinetics Core, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | - Amy Q Wang
- Drug Metabolism and Pharmacokinetics Core, National Center for Advancing Translational Sciences, Rockville, MD, USA
| | | | - Joshua K Park
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Rockville, MD, USA
| | - Charles N Zawatsky
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Rockville, MD, USA
| | - May Christine V Malicdan
- NIH Undiagnosed Diseases Program, UDP Translational Laboratory, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Rockville, MD, USA
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Rockville, MD 20852, USA
| | | | | | - Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, National Institute of Health, Rockville, MD, USA
| | - Xin Xu
- Drug Metabolism and Pharmacokinetics Core, National Center for Advancing Translational Sciences, Rockville, MD, USA.
| |
Collapse
|
4
|
Ramirez-Hincapie S, Birk B, Ternes P, Giri V, Zickgraf FM, Haake V, Herold M, Kamp H, Driemert P, Landsiedel R, Richling E, Funk-Weyer D, van Ravenzwaay B. Application of high throughput in vitro metabolomics for hepatotoxicity mode of action characterization and mechanistic-anchored point of departure derivation: a case study with nitrofurantoin. Arch Toxicol 2023; 97:2903-2917. [PMID: 37665362 PMCID: PMC10504224 DOI: 10.1007/s00204-023-03572-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Omics techniques have been increasingly recognized as promising tools for Next Generation Risk Assessment. Targeted metabolomics offer the advantage of providing readily interpretable mechanistic information about perturbed biological pathways. In this study, a high-throughput LC-MS/MS-based broad targeted metabolomics system was applied to study nitrofurantoin metabolic dynamics over time and concentration and to provide a mechanistic-anchored approach for point of departure (PoD) derivation. Upon nitrofurantoin exposure at five concentrations (7.5 µM, 15 µM, 20 µM, 30 µM and 120 µM) and four time points (3, 6, 24 and 48 h), the intracellular metabolome of HepG2 cells was evaluated. In total, 256 uniquely identified metabolites were measured, annotated, and allocated in 13 different metabolite classes. Principal component analysis (PCA) and univariate statistical analysis showed clear metabolome-based time and concentration effects. Mechanistic information evidenced the differential activation of cellular pathways indicative of early adaptive and hepatotoxic response. At low concentrations, effects were seen mainly in the energy and lipid metabolism, in the mid concentration range, the activation of the antioxidant cellular response was evidenced by increased levels of glutathione (GSH) and metabolites from the de novo GSH synthesis pathway. At the highest concentrations, the depletion of GSH, together with alternations reflective of mitochondrial impairments, were indicative of a hepatotoxic response. Finally, a metabolomics-based PoD was derived by multivariate PCA using the whole set of measured metabolites. This approach allows using the entire dataset and derive PoD that can be mechanistically anchored to established key events. Our results show the suitability of high throughput targeted metabolomics to investigate mechanisms of hepatoxicity and derive point of departures that can be linked to existing adverse outcome pathways and contribute to the development of new ones.
Collapse
Affiliation(s)
| | - Barbara Birk
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | - Varun Giri
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
| | | | | | | | | | | | - Robert Landsiedel
- BASF SE, Experimental Toxicology and Ecology, Ludwigshafen, Germany
- Pharmacy, Pharmacology and Toxicology, Free University of Berlin, Berlin, Germany
| | - Elke Richling
- Food Chemistry and Toxicology, Department of Chemistry, RPTU Kaiserslautern-Landau, Kaiserslautern, Germany
| | | | | |
Collapse
|
5
|
Cai L, Ke M, Wang H, Wu W, Lin R, Huang P, Lin C. Physiologically based pharmacokinetic model combined with reverse dose method to study the nephrotoxic tolerance dose of tacrolimus. Arch Toxicol 2023; 97:2659-2673. [PMID: 37572130 DOI: 10.1007/s00204-023-03576-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Nephrotoxicity is the most common side effect that severely limits the clinical application of tacrolimus (TAC), an immunosuppressive agent used in kidney transplant patients. This study aimed to explore the tolerated dose of nephrotoxicity of TAC in individuals with different CYP3A5 genotypes and liver conditions. We established a human whole-body physiological pharmacokinetic (WB-PBPK) model and validated it using data from previous clinical studies. Following the injection of 1 mg/kg TAC into the tail veins of male rats, we developed a rat PBPK model utilizing the drug concentration-time curve obtained by LC-MS/MS. Next, we converted the established rat PBPK model into the human kidney PBPK model. To establish renal concentrations, the BMCL5 of the in vitro CCK-8 toxicity response curve (drug concentration range: 2-80 mol/L) was extrapolated. To further investigate the acceptable levels of nephrotoxicity for several distinct CYP3A5 genotypes and varied hepatic function populations, oral dosing regimens were extrapolated utilizing in vitro-in vivo extrapolation (IVIVE). The PBPK model indicated the tolerated doses of nephrotoxicity were 0.14-0.185 mg/kg (CYP3A5 expressors) and 0.13-0.155 mg/kg (CYP3A5 non-expressors) in normal healthy subjects and 0.07-0.09 mg/kg (CYP3A5 expressors) and 0.06-0.08 mg/kg (CYP3A5 non-expressors) in patients with mild hepatic insufficiency. Further, patients with moderate hepatic insufficiency tolerated doses of 0.045-0.06 mg/kg (CYP3A5 expressors) and 0.04-0.05 mg/kg (CYP3A5 non-expressors), while in patients with moderate hepatic insufficiency, doses of 0.028-0.04 mg/kg (CYP3A5 expressors) and 0.022-0.03 mg/kg (CYP3A5 non-expressors) were tolerated. Overall, our study highlights the combined usage of the PBPK model and the IVIVE approach as a valuable tool for predicting toxicity tolerated doses of a drug in a specific group.
Collapse
Affiliation(s)
- Limin Cai
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Meng Ke
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Han Wang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Wanhong Wu
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Rongfang Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Pinfang Huang
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China
| | - Cuihong Lin
- Department of Pharmacy, The First Affiliated Hospital of Fujian Medical University, 20 Cha Zhong M. Rd, Fuzhou, 350005, People's Republic of China.
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, People's Republic of China.
| |
Collapse
|
6
|
Widjaja F, Zheng L, Wesseling S, Rietjens IMCM. Physiologically based kinetic modeling of senecionine N-oxide in rats as a new approach methodology to define the effects of dose and endpoint used on relative potency values of pyrrolizidine alkaloid N-oxides. Front Pharmacol 2023; 14:1125146. [PMID: 36937884 PMCID: PMC10017778 DOI: 10.3389/fphar.2023.1125146] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Over 1,000 pyrrolizidine alkaloids (PAs) and their N-oxides (PA-N-oxides) occur in 3% of all flowering plants. PA-N-oxides are toxic when reduced to their parent PAs, which are bioactivated into pyrrole intermediates that generate protein- and DNA-adducts resulting in liver toxicity and carcinogenicity. Literature data for senecionine N-oxide in rats indicate that the relative potency (REP) value of this PA-N-oxide compared to its parent PA senecionine varies with the endpoint used. The first endpoint was the ratio between the area under the concentration-time curve (AUC) for senecionine upon dosing senecionine N-oxide or an equimolar dose of senecionine, while the second endpoint was the ratio between the amount for pyrrole-protein adducts formed under these conditions. This study aimed to investigate the mode of action underlying this endpoint dependent REP value for senecionine N-oxide with physiologically based kinetic (PBK) modeling. Results obtained reveal that limitation of 7-GS-DHP adduct formation due to GSH depletion, resulting in increased pyrrole-protein adduct formation, occurs more likely upon high dose oral PA administration than upon an equimolar dose of PA-N-oxide. At high dose levels, this results in a lower REP value when based on pyrrole-protein adduct levels than when based on PA concentrations. At low dose levels, the difference no longer exists. Altogether, the results of the study show how the REP value for senecionine N-oxide depends on dose and endpoint used, and that PBK modeling provides a way to characterize REP values for PA-N-oxides at realistic low dietary exposure levels, thus reducing the need for animal experiments.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University, Wageningen, Netherlands
| | | | | | | |
Collapse
|
7
|
Mukhtar M, Saleem M, Nazir M, Riaz N, Shafiq N, Saleem H, Tauseef S, Khan S, Ehsan Mazhar M, Bakhsh Tareen R, Habib ur Rahman Mahmood M, Imran Tousif M, Chandra Ojha S. Identification of Pyrrolizidine Alkaloids and Flavonoid Glycosides Through HR-LCMS/MS Analysis, Biological Screening, DFT and Molecular Docking Studies on Heliotropium dasycarpum Ledeb. ARAB J CHEM 2023. [DOI: 10.1016/j.arabjc.2023.104655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
8
|
Widjaja F, Alhejji Y, Yangchen J, Wesseling S, Rietjens IMCM. Physiologically-Based Kinetic Modeling Predicts Similar In Vivo Relative Potency of Senecionine N-Oxide for Rat and Human at Realistic Low Exposure Levels. Mol Nutr Food Res 2023; 67:e2200293. [PMID: 36478522 DOI: 10.1002/mnfr.202200293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 10/30/2022] [Indexed: 12/12/2022]
Abstract
SCOPE This study aims to determine if previously developed physiologically-based kinetic (PBK) model in rat can be modified for senecionine (SEN) and its N-oxide (SENO), and be used to investigate potential species differences between rat and human in relative potency (REP) of the N-oxide relative to the parent pyrrolizidine alkaloid (PA). METHODS AND RESULTS In vitro derived kinetic parameters including the apparent maximum velocities (Vmax ) and Michaelis-Menten constants (Km ) for SENO reduction and SEN clearance are used to define the PBK models. The rat model is validated with published animal data, and the toxicokinetic profiles of SEN from either orally-administered SENO or SEN are simulated. REP values of SENO relative to SEN amount to 0.84 and 0.89 in rat and human, respectively. CONCLUSION The REP value can be dose- and species-dependent, with the values for rat and human being comparable at low realistic exposure scenarios. In summary, PBK modeling serves as a valuable New Approach Methodology (NAM) tool for predicting REP values of PA-N-oxides and may actually result in more accurate REP values for human risk assessment than what would be defined using in vivo animal experiments.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University, PO Box 8000, Wageningen, 6700 EA, The Netherlands
| | - Yasser Alhejji
- Division of Toxicology, Wageningen University, PO Box 8000, Wageningen, 6700 EA, The Netherlands.,Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, 51452, Saudi Arabia
| | - Jamyang Yangchen
- Division of Toxicology, Wageningen University, PO Box 8000, Wageningen, 6700 EA, The Netherlands.,Bhutan Agriculture and Food Regulatory Authority, Ministry of Agriculture and Forests, Thimphu, 11002, Bhutan
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University, PO Box 8000, Wageningen, 6700 EA, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, PO Box 8000, Wageningen, 6700 EA, The Netherlands
| |
Collapse
|
9
|
Dimitrijevic D, Fabian E, Nicol B, Funk-Weyer D, Landsiedel R. Toward Realistic Dosimetry In Vitro: Determining Effective Concentrations of Test Substances in Cell Culture and Their Prediction by an In Silico Mass Balance Model. Chem Res Toxicol 2022; 35:1962-1973. [PMID: 36264934 PMCID: PMC9682521 DOI: 10.1021/acs.chemrestox.2c00128] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Nominal concentrations (CNom) in cell culture media are routinely used to define concentration-effect relationships in the in vitro toxicology. The actual concentration in the medium (CMedium) can be affected by adsorption processes, evaporation, or degradation of chemicals. Therefore, we measured the total and free concentration of 12 chemicals, covering a wide range of lipophilicity (log KOW -0.07-6.84), in the culture medium (CMedium) and cells (CCell) after incubation with Balb/c 3T3 cells for up to 48 h. Measured values were compared to predictions using an as yet unpublished in silico mass balance model that combined relevant equations from similar models published by others. The total CMedium for all chemicals except tamoxifen (TAM) were similar to the CNom. This was attributed to the cellular uptake of TAM and accumulation into lysosomes. The free (i.e., unbound) CMedium for the low/no protein binding chemicals were similar to the CNom, whereas values of all moderately to highly protein-bound chemicals were less than 30% of the CNom. Of the 12 chemicals, the two most hydrophilic chemicals, acetaminophen (APAP) and caffeine (CAF), were the only ones for which the CCell was the same as the CNom. The CCell for all other chemicals tended to increase over time and were all 2- to 274-fold higher than CNom. Measurements of CCytosol, using a digitonin method to release cytosol, compared well with CCell (using a freeze-thaw method) for four chemicals (CAF, APAP, FLU, and KET), indicating that both methods could be used. The mass balance model predicted the total CMedium within 30% of the measured values for 11 chemicals. The free CMedium of all 12 chemicals were predicted within 3-fold of the measured values. There was a poorer prediction of CCell values, with a median overprediction of 3- to 4-fold. In conclusion, while the number of chemicals in the study is limited, it demonstrates the large differences between CNom and total and free CMedium and CCell, which were also relatively well predicted by the mass balance model.
Collapse
Affiliation(s)
- Dunja Dimitrijevic
- Free
University of Berlin, Institute of Pharmacy, Pharmacology and Toxicology, Königin-Luise-Straße
2−4, 14195Berlin, Germany
| | - Eric Fabian
- BASF
SE, Experimental Toxicology and Ecology, Carl-Bosch-Straße 38, 67056Ludwigshafen am Rhein, Germany
| | - Beate Nicol
- Safety
& Environmental Assurance Centre, Unilever
U.K., Sharnbrook, MK44 ILQBedford, United Kingdom
| | - Dorothee Funk-Weyer
- BASF
SE, Experimental Toxicology and Ecology, Carl-Bosch-Straße 38, 67056Ludwigshafen am Rhein, Germany
| | - Robert Landsiedel
- Free
University of Berlin, Institute of Pharmacy, Pharmacology and Toxicology, Königin-Luise-Straße
2−4, 14195Berlin, Germany,BASF
SE, Experimental Toxicology and Ecology, Carl-Bosch-Straße 38, 67056Ludwigshafen am Rhein, Germany,. Fax: +49 621 60-58134
| |
Collapse
|
10
|
Swaters D, van Veen A, van Meurs W, Turner JE, Ritskes-Hoitinga M. A History of Regulatory Animal Testing: What Can We Learn? Altern Lab Anim 2022; 50:322-329. [PMID: 35983829 DOI: 10.1177/02611929221118001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The contemporary pharmaceutical industry is voicing growing concerns about the translatability and reproducibility of animal models. In addition, the usefulness of certain of the required regulatory safety tests in animals is being increasingly questioned. It remains difficult, however, to make the move toward alternative testing methods, not least because of legislative demands. A historical analysis was performed, in order to study how the mandatory animal studies in legislative requirements came about. This article reflects on the role that specific public health disasters played in the creation of (more) regulatory requirements for animal testing. It will show how the regulatory changes prompted by the sulfanilamide elixir disaster in the 1930s and the thalidomide disaster in the early 1960s were based on the belief that extensive animal testing would prevent similar future human health tragedies. As scientists increasingly highlight issues with translatability between non-human animals and humans, the belief that current regulatory requirements ensure safety becomes more difficult to maintain. In addition, it means that some of the regulations now in place require animal tests that do not contribute to the safety of a drug, as shown in a third case study of the court case by Vanda industries against the FDA. We finally argue that regulations should be critically examined and altered where necessary, so that they are no longer a barrier in the transition toward animal-free testing and more human-relevant science.
Collapse
Affiliation(s)
- Doortje Swaters
- Radboud Institute for Culture and History, 6029Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Anne van Veen
- Nijmegen School of Management, 6029Radboud Universiteit Nijmegen, Nijmegen, The Netherlands
| | - Wim van Meurs
- Radboud Institute for Culture and History, 6029Radboud University Nijmegen, Nijmegen, The Netherlands
| | | | - Merel Ritskes-Hoitinga
- SYRCLE, Department for Health Evidence, 6029Radboudumc (Radboud University Medical Center), Nijmegen, The Netherlands.,AUGUST, Department for Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
11
|
Giera DS, Preisitsch M, Brevard H, Nemetz J. Quantitative Removal of Pyrrolizidine Alkaloids from Essential Oils by the Hydrodistillation Step in Their Manufacturing Process. PLANTA MEDICA 2022; 88:538-547. [PMID: 34293805 DOI: 10.1055/a-1534-6928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Pyrrolizidine alkaloids are naturally occurring toxins produced by certain weeds that can, if accidentally co-harvested, contaminate plant-based food, feed, and herbal medicinal products. Focusing on herbal medicinal products, the presence of pyrrolizidine alkaloids is restricted by regulatory prescribed thresholds to assure patient safety. Among the multitude of different herbal active substances utilized in herbal medicinal products, the class of pharmaceutically effective essential oils is considered to exhibit a negligible contribution to pyrrolizidine alkaloid contamination. Within the present investigation, this hypothesis should be scientifically scrutinized. For this purpose, an experimental set-up was chosen that reproduces the typical manufacturing step of hydrodistillation. Essential oils of eucalyptus and lemon were selected exemplarily and spiked with 3 representative pyrrolizidine alkaloids (retrorsine, retrorsine-N-oxide, and lycopsamine), whereupon hydrodistillation was performed. Analysis of the resulting distillates by LC-MS/MS proved that artificially added pyrrolizidine alkaloids were removed completely. Moreover, quantitative pyrrolizidine alkaloid recovery in the aqueous phases was observed. Hence, it was experimentally confirmed that herbal medicinal products employing hydrodistilled essential oils of pharmaceutical quality are intrinsically free of pyrrolizidine alkaloids due to the particularities of their manufacturing process. Furthermore, it can be concluded from theoretical considerations that essential oils produced by cold pressing have a negligible risk of carrying pyrrolizidine alkaloid contamination. Our findings provide a strong indication that the requirement for analytical pyrrolizidine alkaloid testing of essential oils for pharmaceutical use should be fundamentally reconsidered.
Collapse
Affiliation(s)
- David S Giera
- G. Pohl-Boskamp GmbH & Co. KG, Hohenlockstedt, Germany
| | | | | | - Jörn Nemetz
- G. Pohl-Boskamp GmbH & Co. KG, Hohenlockstedt, Germany
| |
Collapse
|
12
|
Noorlander A, Zhang M, van Ravenzwaay B, Rietjens IMCM. Use of physiologically based kinetic modeling-facilitated reverse dosimetry to predict in vivo acute toxicity of tetrodotoxin in rodents. Toxicol Sci 2022; 187:127-138. [PMID: 35218365 PMCID: PMC9041554 DOI: 10.1093/toxsci/kfac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
In this study, the ability of a new in vitro/in silico quantitative in vitro–in vivo extrapolation (QIVIVE) methodology was assessed to predict the in vivo neurotoxicity of tetrodotoxin (TTX) in rodents. In vitro concentration–response data of TTX obtained in a multielectrode array assay with primary rat neonatal cortical cells and in an effect study with mouse neuro-2a cells were quantitatively extrapolated into in vivo dose–response data, using newly developed physiologically based kinetic (PBK) models for TTX in rats and mice. Incorporating a kidney compartment accounting for active renal excretion in the PBK models proved to be essential for its performance. To evaluate the predictions, QIVIVE-derived dose–response data were compared with in vivo data on neurotoxicity in rats and mice upon oral and parenteral dosing. The results revealed that for both rats and mice the predicted dose–response data matched the data from available in vivo studies well. It is concluded that PBK modeling-based reserve dosimetry of in vitro TTX effect data can adequately predict the in vivo neurotoxicity of TTX in rodents, providing a novel proof-of-principle for this methodology.
Collapse
Affiliation(s)
- Annelies Noorlander
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Mengying Zhang
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Bennard van Ravenzwaay
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands.,Experimental Toxicology and Ecology, BASF SE, Z 470, Ludwigshafen, 67056, Germany
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| |
Collapse
|
13
|
Widjaja F, Alhejji Y, Rietjens IMCM. The Role of Kinetics as Key Determinant in Toxicity of Pyrrolizidine Alkaloids and Their N-Oxides. PLANTA MEDICA 2022; 88:130-143. [PMID: 34741297 PMCID: PMC8807025 DOI: 10.1055/a-1582-9794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are a large group of plant constituents of which especially the 1,2- unsaturated PAs raise a concern because of their liver toxicity and potential genotoxic carcinogenicity. This toxicity of PAs depends on their kinetics. Differences in absorption, distribution, metabolism, and excretion (ADME) characteristics of PAs may substantially alter the relative toxicity of PAs. As a result, kinetics will also affect relative potency (REP) values. The present review summarizes the current state-of-the art on PA kinetics and resulting consequences for toxicity and illustrates how physiologically-based kinetic (PBK) modelling can be applied to take kinetics into account when defining the relative differences in toxicity between PAs in the in vivo situation. We conclude that toxicokinetics play an important role in the overall toxicity of pyrrolizidine alkaloids. and that kinetics should therefore be considered when defining REP values for combined risk assessment. New approach methodologies (NAMs) can be of use to quantify these kinetic differences between PAs and their N-oxides, thus contributing to the 3Rs (Replacement, Reduction and Refinement) in animal studies.
Collapse
Affiliation(s)
- Frances Widjaja
- Division of Toxicology, Wageningen University and Research, The Netherlands
| | - Yasser Alhejji
- Division of Toxicology, Wageningen University and Research, The Netherlands
- Department of Food Science and Human Nutrition, College of Agriculture and Veterinary Medicine, Qassim University, Buraydah, Saudi Arabia
| | | |
Collapse
|
14
|
Wiesner J. Regulatory Perspectives of Pyrrolizidine Alkaloid Contamination in Herbal Medicinal Products. PLANTA MEDICA 2022; 88:118-124. [PMID: 34169489 DOI: 10.1055/a-1494-1363] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The toxicity of plants containing certain pyrrolizidine alkaloids has long been recognized in grazing animals and humans. Genotoxicity and carcinogenicity data from in vitro and in vivo (animal) studies were published over the last few decades for some of the 1,2-unsaturated pyrrolizidine alkaloids, leading to regulatory action on herbal medicinal products with pyrrolizidine alkaloid-containing plants more than 30 years ago. In recent years, it has become evident that in addition to herbal medicinal products containing pyrrolizidine alkaloid-containing plants, these products may also contain pyrrolizidine alkaloids without actually including pyrrolizidine alkaloid-containing plants. This is explained by contamination by accessory herbs (weeds). The national competent authorities of the European member states and the European Medicines Agency, in this case, the Committee on Herbal Medicinal Products, reacted to these findings by setting limits for all herbal medicinal products. This review article will briefly discuss the data leading to the establishment of thresholds and the regulatory developments and consequences, as well as the current discussions and research in this area.
Collapse
|
15
|
Physiologically based kinetic modelling predicts the in vivo relative potency of riddelliine N-oxide compared to riddelliine in rat to be dose dependent. Arch Toxicol 2021; 96:135-151. [PMID: 34669010 PMCID: PMC8748370 DOI: 10.1007/s00204-021-03179-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022]
Abstract
Pyrrolizidine alkaloids (PAs) are toxic plant constituents occurring often in their N-oxide form. This raises the question on the relative potency (REP) values of PA-N-oxides compared to the corresponding parent PAs. The present study aims to quantify the in vivo REP value of riddelliine N-oxide compared to riddelliine using physiologically based kinetic (PBK) modelling, taking into account that the toxicity of riddelliine N-oxide depends on its conversion to riddelliine by intestinal microbiota and in the liver. The models predicted a lower Cmax and higher Tmax for the blood concentration of riddelliine upon oral administration of riddelliine N-oxide compared to the Cmax and Tmax predicted for an equimolar oral dose of riddelliine. Comparison of the area under the riddelliine concentration–time curve (AUCRID) obtained upon dosing either the N-oxide or riddelliine itself revealed a ratio of 0.67, which reflects the in vivo REP for riddelliine N-oxide compared to riddelliine, and appeared to closely match the REP value derived from available in vivo data. The models also predicted that the REP value will decrease with increasing dose level, because of saturation of riddelliine N-oxide reduction by the intestinal microbiota and of riddelliine clearance by the liver. It is concluded that PBK modeling provides a way to define in vivo REP values of PA-N-oxides as compared to their parent PAs, without a need for animal experiments.
Collapse
|
16
|
Ma J, Li M, Li N, Chan WY, Lin G. Pyrrolizidine Alkaloid-Induced Hepatotoxicity Associated with the Formation of Reactive Metabolite-Derived Pyrrole-Protein Adducts. Toxins (Basel) 2021; 13:723. [PMID: 34679016 PMCID: PMC8540779 DOI: 10.3390/toxins13100723] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/19/2022] Open
Abstract
Pyrrolizidine alkaloids (PAs) with 1,2-unsaturated necine base are hepatotoxic phytotoxins. Acute PA intoxication is initiated by the formation of adducts between PA-derived reactive pyrrolic metabolites with cellular proteins. The present study aimed to investigate the correlation between the formation of hepatic pyrrole-protein adducts and occurrence of PA-induced liver injury (PA-ILI), and to further explore the use of such adducts for rapidly screening the hepatotoxic potency of natural products which contain PAs. Aqueous extracts of Crotalaria sessiliflora (containing one PA: monocrotaline) and Gynura japonica (containing two PAs: senecionine and seneciphylline) were orally administered to rats at different doses for 24 h to investigate PA-ILI. Serum alanine aminotransferase (ALT) activity, hepatic glutathione (GSH) level, and liver histological changes of the treated rats were evaluated to assess the severity of PA-ILI. The levels of pyrrole-protein adducts formed in the rats' livers were determined by a well-established spectrophotometric method. The biological and histological results showed a dose-dependent hepatotoxicity with significantly different toxic severity among groups of rats treated with herbal extracts containing different PAs. Both serum ALT activity and the amount of hepatic pyrrole-protein adducts increased in a dose-dependent manner. Moreover, the elevation of ALT activity correlated well with the formation of hepatic pyrrole-protein adducts, regardless of the structures of different PAs. The findings revealed that the formation of hepatic pyrrole-protein adducts-which directly correlated with the elevation of serum ALT activity-was a common insult leading to PA-ILI, suggesting a potential for using pyrrole-protein adducts to screen hepatotoxicity and rank PA-containing natural products, which generally contain multiple PAs with different structures.
Collapse
Affiliation(s)
- Jiang Ma
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 4054577, China; (J.M.); (M.L.); (N.L.); (W.Y.C.)
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Mi Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 4054577, China; (J.M.); (M.L.); (N.L.); (W.Y.C.)
| | - Na Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 4054577, China; (J.M.); (M.L.); (N.L.); (W.Y.C.)
| | - Wood Yee Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 4054577, China; (J.M.); (M.L.); (N.L.); (W.Y.C.)
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 4054577, China; (J.M.); (M.L.); (N.L.); (W.Y.C.)
| |
Collapse
|
17
|
Zhang M, van Ravenzwaay B, Rietjens IMCM. Development of a Generic Physiologically Based Kinetic Model to Predict In Vivo Uterotrophic Responses Induced by Estrogenic Chemicals in Rats Based on In Vitro Bioassays. Toxicol Sci 2021; 173:19-31. [PMID: 31626307 PMCID: PMC9186316 DOI: 10.1093/toxsci/kfz216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The present study assessed the potential of a generic physiologically based kinetic (PBK) model to convert in vitro data for estrogenicity to predict the in vivo uterotrophic response in rats for diethylstibestrol (DES), ethinylestradiol (EE2), genistein (GEN), coumestrol (COU), and methoxychlor (MXC). PBK models were developed using a generic approach and in vitro concentration-response data from the MCF-7 proliferation assay and the yeast estrogen screening assay were translated into in vivo dose-response data. Benchmark dose analysis was performed on the predicted data and available in vivo uterotrophic data to evaluate the model predictions. The results reveal that the developed generic PBK model adequate defines the in vivo kinetics of the estrogens. The predicted dose-response data of DES, EE2, GEN, COU, and MXC matched the reported in vivo uterus weight response in a qualitative way, whereas the quantitative comparison was somewhat hampered by the variability in both in vitro and in vivo data. From a safety perspective, the predictions based on the MCF-7 proliferation assay would best guarantee a safe point of departure for further risk assessment although it may be conservative. The current study indicates the feasibility of using a combination of in vitro toxicity data and a generic PBK model to predict the relative in vivo uterotrophic response for estrogenic chemicals.
Collapse
Affiliation(s)
- Mengying Zhang
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, The Netherlands
| | - Bennard van Ravenzwaay
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, The Netherlands.,Experimental Toxicology and Ecology, BASF SE, Z 470, Ludwigshafen 67056, Germany
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, The Netherlands
| |
Collapse
|
18
|
Zhao S, Wesseling S, Spenkelink B, Rietjens IMCM. Physiologically based kinetic modelling based prediction of in vivo rat and human acetylcholinesterase (AChE) inhibition upon exposure to diazinon. Arch Toxicol 2021; 95:1573-1593. [PMID: 33715020 PMCID: PMC8113213 DOI: 10.1007/s00204-021-03015-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/25/2021] [Indexed: 01/30/2023]
Abstract
The present study predicts in vivo human and rat red blood cell (RBC) acetylcholinesterase (AChE) inhibition upon diazinon (DZN) exposure using physiological based kinetic (PBK) modelling-facilitated reverse dosimetry. Due to the fact that both DZN and its oxon metabolite diazoxon (DZO) can inhibit AChE, a toxic equivalency factor (TEF) was included in the PBK model to combine the effect of DZN and DZO when predicting in vivo AChE inhibition. The PBK models were defined based on kinetic constants derived from in vitro incubations with liver fractions or plasma of rat and human, and were used to translate in vitro concentration-response curves for AChE inhibition obtained in the current study to predicted in vivo dose-response curves. The predicted dose-response curves for rat matched available in vivo data on AChE inhibition, and the benchmark dose lower confidence limits for 10% inhibition (BMDL10 values) were in line with the reported BMDL10 values. Humans were predicted to be 6-fold more sensitive than rats in terms of AChE inhibition, mainly because of inter-species differences in toxicokinetics. It is concluded that the TEF-coded DZN PBK model combined with quantitative in vitro to in vivo extrapolation (QIVIVE) provides an adequate approach to predict RBC AChE inhibition upon acute oral DZN exposure, and can provide an alternative testing strategy for derivation of a point of departure (POD) in risk assessment.
Collapse
Affiliation(s)
- Shensheng Zhao
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Bert Spenkelink
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|
19
|
Henneberger L, Huchthausen J, Wojtysiak N, Escher BI. Quantitative In Vitro-to- In Vivo Extrapolation: Nominal versus Freely Dissolved Concentration. Chem Res Toxicol 2021; 34:1175-1182. [PMID: 33759508 DOI: 10.1021/acs.chemrestox.1c00037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Discussions are ongoing on which dose metric should be used for quantitative in vitro-to-in vivo extrapolation (QIVIVE) of in vitro bioassay data. The nominal concentration of the test chemicals is most commonly used and easily accessible, while the concentration freely dissolved in the assay medium is considered to better reflect the bioavailable concentration but is tedious to measure. The aim of this study was to elucidate how much QIVIVE results will differ when using either nominal or freely dissolved concentrations. QIVIVEnom and QIVIVEfree ratios, that is, the ratios of plasma concentrations divided by in vitro effect concentrations, were calculated for 10 pharmaceuticals using previously published nominal and freely dissolved effect concentrations for the activation of the peroxisome proliferator-activated receptor gamma (PPARγ) and the activation of oxidative stress response. The QIVIVEnom ratios were higher than QIVIVEfree ratios by up to a factor of 60. The risk of in vivo effects was classified as being high or low for four chemicals using the QIVIVEnom and for three chemicals using QIVIVEfree ratios. Unambiguous classification was possible for nine chemicals by combining the QIVIVEnom or QIVIVEfree ratios with the respective specificity ratios (SRnom or SRfree) of the in vitro effect data, which helps to identify whether the specific effect was influenced by cytotoxicity. QIVIVEfree models should be preferred as they account for differences in bioavailability between in vitro and in vivo, but QIVIVEnom may still be useful for screening the effects of large numbers of chemicals because it is generally more conservative. The use of SR of the in vitro effect data as a second classification factor is recommended for QIVIVEnom and QIVIVEfree models because a clearer picture can be obtained with respect to the likelihood that a biological effect will occur and that it is not caused by nonspecific cytotoxicity.
Collapse
Affiliation(s)
- Luise Henneberger
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstr. 15, 04318 Leipzig, Germany
| | - Julia Huchthausen
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstr. 15, 04318 Leipzig, Germany
| | - Niklas Wojtysiak
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstr. 15, 04318 Leipzig, Germany
| | - Beate I Escher
- Department of Cell Toxicology, Helmholtz Centre for Environmental Research - UFZ, Permoserstr. 15, 04318 Leipzig, Germany.,Environmental Toxicology, Center for Applied Geoscience, Eberhard Karls University Tübingen, Schnarrenbergstr. 94-96, 72076 Tübingen, Germany
| |
Collapse
|
20
|
Incorporating renal excretion via the OCT2 transporter in physiologically based kinetic modelling to predict in vivo kinetics of mepiquat in rat. Toxicol Lett 2021; 343:34-43. [PMID: 33639197 DOI: 10.1016/j.toxlet.2021.02.013] [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: 12/11/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 01/14/2023]
Abstract
The present study aimed at incorporating active renal excretion via the organic cation transporter 2 (OCT2) into a generic rat physiologically based kinetic (PBK) model using an in vitro human renal proximal tubular epithelial cell line (SA7K) and mepiquat chloride (MQ) as the model compound. The Vmax (10.5 pmol/min/mg protein) and Km (20.6 μM) of OCT2 transport of MQ were determined by concentration-dependent uptake in SA7K cells using doxepin as inhibitor. PBK model predictions incorporating these values in the PBK model were 6.7-8.4-fold different from the reported in vivo data on the blood concentration of MQ in rat. Applying an overall scaling factor that also corrects for potential differences in OCT2 activity in the SA7K cells and in vivo kidney cortex and species differences resulted in adequate predictions for in vivo kinetics of MQ in rat (2.3-3.2-fold). The results indicate that using SA7K cells to define PBK parameters for active renal OCT2 mediated excretion with adequate scaling enables incorporation of renal excretion via the OCT2 transporter in PBK modelling to predict in vivo kinetics of mepiquat in rat. This study demonstrates a proof-of-principle on how to include active renal excretion into generic PBK models.
Collapse
|
21
|
Noorlander A, Fabian E, van Ravenzwaay B, Rietjens IMCM. Novel testing strategy for prediction of rat biliary excretion of intravenously administered estradiol-17β glucuronide. Arch Toxicol 2021; 95:91-102. [PMID: 33159584 PMCID: PMC7811516 DOI: 10.1007/s00204-020-02908-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/10/2020] [Indexed: 10/31/2022]
Abstract
The aim of the present study was to develop a generic rat physiologically based kinetic (PBK) model that includes a novel testing strategy where active biliary excretion is incorporated using estradiol-17β glucuronide (E217βG) as the model substance. A major challenge was the definition of the scaling factor for the in vitro to in vivo conversion of the PBK-model parameter Vmax. In vitro values for the Vmax and Km for transport of E217βG were found in the literature in four different studies based on experiments with primary rat hepatocytes. The required scaling factor was defined based on fitting the PBK model-based predicted values to reported experimental data on E217βG blood levels and cumulative biliary E217βG excretion. This resulted in a scaling factor of 129 mg protein/g liver. With this scaling factor the PBK model predicted the in vivo data for blood and cumulative biliary E217βG levels with on average of less than 1.8-fold deviation. The study provides a proof of principle on how biliary excretion can be included in a generic PBK model using primary hepatocytes to define the kinetic parameters that describe the biliary excretion.
Collapse
Affiliation(s)
- Annelies Noorlander
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands.
| | - Eric Fabian
- Experimental Toxicology and Ecology, BASF SE, Ludwigshafen, Germany
| | | | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|
22
|
He Y, Zhu L, Ma J, Wong L, Zhao Z, Ye Y, Fu PP, Lin G. Comprehensive investigation and risk study on pyrrolizidine alkaloid contamination in Chinese retail honey. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 267:115542. [PMID: 33254676 DOI: 10.1016/j.envpol.2020.115542] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/20/2020] [Accepted: 08/24/2020] [Indexed: 06/12/2023]
Abstract
Pyrrolizidine alkaloids (PAs) are common phytotoxins. We performed the first comprehensive investigation on PA contamination in Chinese honeys. LC-MS analysis revealed that 58% of 255 honey samples purchased from 17 regions across Mainland China and Taiwan contained PAs with total content ranging over 0.2-281.1 μg/kg. Monocrotaline (from Crotalaria spp), a PA never found in honey in other regions, together with echimidine (Echium plantagineum) and lycopsamine (from Senecio spp.), were three predominant PAs in PA-contaminated Chinese honeys. Further, PAs present in honeys were found to have geographically distinct pattern, indicating possible control of such contamination in future honey production. Moreover, we proposed a new risk estimation approach, which considered both content and toxic potency of individual PAs in honeys, and found that 12% of the PA-contaminated Chinese honeys tested might pose potential health risk. This study revealed a high prevalence and potential health risk of PA contamination in Chinese honeys.
Collapse
Affiliation(s)
- Yisheng He
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines between the Chinese University of Hong Kong and Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Lin Zhu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines between the Chinese University of Hong Kong and Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Jiang Ma
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines between the Chinese University of Hong Kong and Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Lailai Wong
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Zhongzhen Zhao
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yang Ye
- Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines between the Chinese University of Hong Kong and Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China
| | - Peter P Fu
- National Center for Toxicological Research, Jefferson, AR 72079, United States
| | - Ge Lin
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Joint Research Laboratory for Promoting Globalization of Traditional Chinese Medicines between the Chinese University of Hong Kong and Shanghai Institute of Materia Medica, Chinese Academy of Sciences, China.
| |
Collapse
|
23
|
Gilbert‐Sandoval I, Wesseling S, Rietjens IMCM. Predicting the Acute Liver Toxicity of Aflatoxin B1 in Rats and Humans by an In Vitro-In Silico Testing Strategy. Mol Nutr Food Res 2020; 64:e2000063. [PMID: 32421213 PMCID: PMC7379280 DOI: 10.1002/mnfr.202000063] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/01/2020] [Indexed: 11/14/2022]
Abstract
SCOPE High-level exposure to aflatoxin B1 (AFB1) is known to cause acute liver damage and fatality in animals and humans. The intakes actually causing this acute toxicity have so far been estimated based on AFB1 levels in contaminated foods or biomarkers in serum. The aim of the present study is to predict the doses causing acute liver toxicity of AFB1 in rats and humans by an in vitro-in silico testing strategy. METHODS AND RESULTS Physiologically based kinetic (PBK) models for AFB1 in rats and humans are developed. The models are used to translate in vitro concentration-response curves for cytotoxicity in primary rat and human hepatocytes to in vivo dose-response curves using reverse dosimetry. From these data, the dose levels at which toxicity would be expected are obtained and compared to toxic dose levels from available rat and human case studies on AFB1 toxicity. The results show that the in vitro-in silico testing strategy can predict dose levels causing acute toxicity of AFB1 in rats and human. CONCLUSIONS Quantitative in vitro in vivo extrapolation (QIVIVE) using PBK modeling-based reverse dosimetry can predict AFB1 doses that cause acute liver toxicity in rats and human.
Collapse
Affiliation(s)
- Ixchel Gilbert‐Sandoval
- Division of ToxicologyWageningen University and ResearchStippeneng 4Wageningen6708 WEThe Netherlands
| | - Sebastiaan Wesseling
- Division of ToxicologyWageningen University and ResearchStippeneng 4Wageningen6708 WEThe Netherlands
| | - Ivonne M. C. M. Rietjens
- Division of ToxicologyWageningen University and ResearchStippeneng 4Wageningen6708 WEThe Netherlands
| |
Collapse
|
24
|
Suparmi S, Wesseling S, Rietjens IMCM. Monocrotaline-induced liver toxicity in rat predicted by a combined in vitro physiologically based kinetic modeling approach. Arch Toxicol 2020; 94:3281-3295. [PMID: 32518961 PMCID: PMC7415757 DOI: 10.1007/s00204-020-02798-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 05/28/2020] [Indexed: 01/03/2023]
Abstract
The aim of the present study was to use an in vitro–in silico approach to predict the in vivo acute liver toxicity of monocrotaline and to characterize the influence of its metabolism on its relative toxic potency compared to lasiocarpine and riddelliine. In the absence of data on acute liver toxicity of monocrotaline upon oral exposure, the predicted dose–response curve for acute liver toxicity in rats and the resulting benchmark dose lower and upper confidence limits for 10% effect (BMDL10 and BMDU10) were compared to data obtained in studies with intraperitoneal or subcutaneous dosing regimens. This indicated the predicted BMDL10 value to be in line with the no-observed-adverse-effect levels (NOAELs) derived from availabe in vivo studies. The predicted BMDL10–BMDU10 of 1.1–4.9 mg/kg bw/day also matched the oral dose range of 1–3 mg PA/kg bw/day at which adverse effects in human are reported. A comparison to the oral toxicity of the related pyrrolizidine alkaloids (PAs) lasiocarpine and riddelliine revealed that, although in the rat hepatocytes monocrotaline was less toxic than lasiocarpine and riddelliine, due to its relatively inefficient clearance, its in vivo acute liver toxicity was predicted to be comparable. It is concluded that the combined in vitro-PBK modeling approach can provide insight in monocrotaline-induced acute liver toxicity in rats, thereby filling existing gaps in the database on PA toxicity. Furthermore, the results reveal that the kinetic and metabolic properties of PAs can vary substantially and should be taken into account when considering differences in relative potency between different PAs.
Collapse
Affiliation(s)
- Suparmi Suparmi
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands. .,Department of Biology, Faculty of Medicine, Universitas Islam Sultan Agung, Jl. Raya Kaligawe KM 4, Semarang, 50112, Indonesia.
| | - Sebastiaan Wesseling
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, The Netherlands
| |
Collapse
|
25
|
Bolt HM, Hengstler JG. The rapid development of computational toxicology. Arch Toxicol 2020; 94:1371-1372. [PMID: 32382955 PMCID: PMC7261728 DOI: 10.1007/s00204-020-02768-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 04/28/2020] [Indexed: 11/15/2022]
Affiliation(s)
- Hermann M Bolt
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystr. 67, 44139, Dortmund, Germany.
| | - Jan G Hengstler
- Department of Toxicology, Leibniz Research Centre for Working Environment and Human Factors at TU Dortmund (IfADo), Ardeystr. 67, 44139, Dortmund, Germany
| |
Collapse
|
26
|
Wang Q, Spenkelink B, Boonpawa R, Rietjens IMCM, Beekmann K. Use of Physiologically Based Kinetic Modeling to Predict Rat Gut Microbial Metabolism of the Isoflavone Daidzein to S-Equol and Its Consequences for ERα Activation. Mol Nutr Food Res 2020; 64:e1900912. [PMID: 32027771 PMCID: PMC7154660 DOI: 10.1002/mnfr.201900912] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/06/2019] [Indexed: 12/13/2022]
Abstract
SCOPE To predict gut microbial metabolism of xenobiotics and the resulting plasma concentrations of metabolites formed, an in vitro-in silico-based testing strategy is developed using the isoflavone daidzein and its gut microbial metabolite S-equol as model compounds. METHODS AND RESULTS Anaerobic rat fecal incubations are optimized and performed to derive the apparent maximum velocities (Vmax ) and Michaelis-Menten constants (Km ) for gut microbial conversion of daidzein to dihydrodaidzein, S-equol, and O-desmethylangolensin, which are input as parameters for a physiologically based kinetic (PBK) model. The inclusion of gut microbiota in the PBK model allows prediction of S-equol concentrations and slightly reduced predicted maximal daidzein concentrations from 2.19 to 2.16 µm. The resulting predicted concentrations of daidzein and S-equol are comparable to in vivo concentrations reported. CONCLUSION The optimized in vitro approach to quantify kinetics for gut microbial conversions, and the newly developed PBK model for rats that includes gut microbial metabolism, provide a unique tool to predict the in vivo consequences of daidzein microbial metabolism for systemic exposure of the host to daidzein and its metabolite S-equol. The predictions reveal a dominant role for daidzein in ERα-mediated estrogenicity despite the higher estrogenic potency of its microbial metabolite S-equol.
Collapse
Affiliation(s)
- Qianrui Wang
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
| | - Bert Spenkelink
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
| | - Rungnapa Boonpawa
- Faculty of Natural Resources and Agro‐IndustryKasetsart University Chalermphrakiat Sakon Nakhon Province CampusSakon Nakhon47000Thailand
| | | | - Karsten Beekmann
- Division of ToxicologyWageningen University and ResearchWageningen6708 WEThe Netherlands
- Present address:
Wageningen Food Safety ResearchP. O. Box 2306700 AEWageningenThe Netherlands
| |
Collapse
|
27
|
Integrating physiologically based kinetic (PBK) and Monte Carlo modelling to predict inter-individual and inter-ethnic variation in bioactivation and liver toxicity of lasiocarpine. Arch Toxicol 2019; 93:2943-2960. [PMID: 31511935 DOI: 10.1007/s00204-019-02563-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/02/2019] [Indexed: 10/26/2022]
Abstract
The aim of the present study was to predict the effect of inter-individual and inter-ethnic human kinetic variation on the sensitivity towards acute liver toxicity of lasiocarpine in the Chinese and the Caucasian population, and to derive chemical specific adjustment factors (CSAFs) by integrating variation in the in vitro kinetic constants Vmax and Km, physiologically based kinetic (PBK) modelling and Monte Carlo simulation. CSAFs were derived covering the 90th and 99th percentile of the population distribution of pyrrole glutathione adduct (7-GS-DHP) formation, reflecting bioactivation. The results revealed that in the Chinese population, as compared to the Caucasian population, the predicted 7-GS-DHP formation at the geometric mean, the 90th and the 99th percentile were 2.1-, 3.3- and 4.3-fold lower respectively. The CSAFs obtained using the 99th percentile values were 8.3, 17.0 and 19.5 in the Chinese, the Caucasian population and the two populations combined, respectively, while the CSAFs were generally 3.0-fold lower at the 90th percentile. These results indicate that when considering the formation of 7-GS-DHP the Caucasian population may be more sensitive towards acute liver toxicity of lasiocarpine, and further point out that the default safety factor of 3.16 for inter-individual human kinetic differences may not be sufficiently protective. Altogether, the results obtained demonstrate that integrating PBK modelling with Monte Carlo simulations using human in vitro data is a powerful strategy to quantify inter-individual variations in kinetics, and can be used to refine the human risk assessment of pyrrolizidine alkaloids.
Collapse
|
28
|
Ning J, Chen L, Rietjens IM. Role of toxicokinetics and alternative testing strategies in pyrrolizidine alkaloid toxicity and risk assessment; state-of-the-art and future perspectives. Food Chem Toxicol 2019; 131:110572. [DOI: 10.1016/j.fct.2019.110572] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/05/2019] [Accepted: 06/07/2019] [Indexed: 01/31/2023]
|
29
|
Prediction of in vivo genotoxicity of lasiocarpine and riddelliine in rat liver using a combined in vitro-physiologically based kinetic modelling-facilitated reverse dosimetry approach. Arch Toxicol 2019; 93:2385-2395. [PMID: 31289892 DOI: 10.1007/s00204-019-02515-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022]
Abstract
Pyrrolizidine alkaloids (PAs) are naturally occurring genotoxic compounds, and PA-containing plants can pose a risk to humans through contaminated food sources and herbal products. Upon metabolic activation, PAs can form DNA adducts, DNA and protein cross links, chromosomal aberrations, micronuclei, and DNA double-strand breaks. These genotoxic effects may induce gene mutations and play a role in the carcinogenesis of PAs. This study aims to predict in vivo genotoxicity for two well-studied PAs, lasiocarpine and riddelliine, in rat using in vitro genotoxicity data and physiologically based kinetic (PBK) modelling-based reverse dosimetry. The phosphorylation of histone protein H2AX was used as a quantitative surrogate endpoint for in vitro genotoxicity of lasiocarpine and riddelliine in primary rat hepatocytes and human HepaRG cells. The in vitro concentration-response curves obtained from primary rat hepatocytes were subsequently converted to in vivo dose-response curves from which points of departure (PoDs) were derived that were compared to available in vivo genotoxicity data. The results showed that the predicted PoDs for lasiocarpine and riddelliine were comparable to in vivo genotoxicity data. It is concluded that this quantitative in vitro-in silico approach provides a method to predict in vivo genotoxicity for the large number of PAs for which in vivo genotoxicity data are lacking by integrating in vitro genotoxicity assays with PBK modelling-facilitated reverse dosimetry.
Collapse
|
30
|
Determination of genotoxic potencies of pyrrolizidine alkaloids in HepaRG cells using the γH2AX assay. Food Chem Toxicol 2019; 131:110532. [PMID: 31154085 DOI: 10.1016/j.fct.2019.05.040] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/23/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
Pyrrolizidine alkaloids (PAs) are secondary metabolites from plants that have been found in substantial amounts in herbal supplements, infusions and teas. Several PAs cause cancer in animal bioassays, mediated via a genotoxic mode of action, but for the majority of the PAs, carcinogenicity data are lacking. It is assumed in the risk assessment that all PAs have the same potency as riddelliine, which is considered to be one of the most potent carcinogenic PAs in rats. This may overestimate the risks, since many PAs are expected to have lower potencies. In this study we determined the concentration-dependent genotoxicity of 37 PAs representing different chemical classes using the γH2AX in cell western assay in HepaRG human liver cells. Based on these in vitro data, PAs were grouped into different potency classes. The group with the highest potency consists particularly of open diester PAs and cyclic diester PAs (including riddelliine). The group of the least potent or non-active PAs includes the monoester PAs, non-esterified necine bases, PA N-oxides, and the unsaturated PA trachelanthamine. This study reveals differences in in vitro genotoxic potencies of PAs, supporting that the assumption that all PAs have a similar potency as riddelliine is rather conservative.
Collapse
|
31
|
|
32
|
Rietjens IMCM, Ning J, Chen L, Wesseling S, Strikwold M, Louisse J. Selecting the dose metric in reverse dosimetry based QIVIVE. Arch Toxicol 2019; 93:1467-1469. [DOI: 10.1007/s00204-019-02438-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 04/03/2019] [Indexed: 12/23/2022]
|