1
|
Duarte P, Sanchez-Porro FJ, Crisman E, Cores Á, Jiménez I, Cuadrado A, Menéndez JC, León R. Network-Based Drug Optimization toward the Treatment of Parkinson's Disease: NRF2, MAO-B, Oxidative Stress, and Chronic Neuroinflammation. J Med Chem 2025. [PMID: 39818855 DOI: 10.1021/acs.jmedchem.4c02659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Parkinson's disease (PD), the second most common neurodegenerative disorder, affects around 10 million people worldwide. It is a multifactorial disease marked by dopaminergic neuron loss with oxidative stress (OS) and neuroinflammation as key pathological drivers. Current treatments focus on dopamine replacement and are symptomatic, underscoring the urgent need for disease-modifying therapies. Here, we present a novel class of dual MAO-B inhibitors and NRF2 inducers with neuroprotective properties in in vitro PD models. Through an optimization program, we enhanced their MAO-B inhibitory potency, selectivity, and NRF2 induction capacity while achieving favorable pharmacokinetic profiles. Virtual library screening identified two core derivatives, leading to the development of compound 11, which exhibited potent anti-inflammatory and neuroprotective activity in OS-related in vitro models. Compound 11 also demonstrated high liver microsomal stability and favorable pharmacokinetics in mice, making it a promising candidate for further investigation as a potential PD therapy.
Collapse
Affiliation(s)
- Pablo Duarte
- Consejo Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| | - Francisco J Sanchez-Porro
- Consejo Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| | - Enrique Crisman
- Consejo Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
- Fundación Teófilo Hernando para la I+D del Medicamento, Las Rozas, 28290 Madrid, Spain
| | - Ángel Cores
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Irene Jiménez
- Consejo Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
- Fundación Teófilo Hernando para la I+D del Medicamento, Las Rozas, 28290 Madrid, Spain
| | - Antonio Cuadrado
- Instituto de Investigación Sanitaria La Paz (IdiPaz) and Departamento de Bioquímica, Facultad de Medicina, UAM, Instituto de Investigaciones Biomédicas "Alberto Sols" UAM-CSIC, 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII, 28029 Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040 Madrid, Spain
| | - Rafael León
- Consejo Superior de Investigaciones Científicas (IQM-CSIC), Instituto de Química Médica, 28006 Madrid, Spain
| |
Collapse
|
2
|
Dulsat J, Puig de la Bellacasa R, Borrell JI. The Use of a Penta-Deuterophenyl Substituent to Improve the Metabolic Stability of a Tyrosine Kinase Inhibitor. Molecules 2024; 29:6042. [PMID: 39770130 PMCID: PMC11679266 DOI: 10.3390/molecules29246042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
In cases in which a rapid metabolism is the cause of an unfavorable pharmacokinetic profile, it is important to determine the Sites of Metabolism (SoMs) of a molecule to introduce the necessary modifications to improve the stability of the compound. The substitution of hydrogen atoms by deuterium atoms has been proposed to ameliorate such properties due to the greater stability of the C-D bonds. IQS016, bearing a 2-phenylamino substituent, is a compound previously described by our group with good biological activity as a discoidin domain receptor (DDR2) inhibitor but suffers from low metabolic stability determined in a test with rat-liver microsomes (less than 50% of the initial compound after 60 min). We have obtained the corresponding 2-(penta-deuterophenyl) analog (IQS016-d5) from aniline-2,3,4,5,6-d5 showing that it has a better metabolic stability than IQS016 and a higher inhibitory effect on isolated tyrosine kinase receptors but not a better 2D in vitro effect.
Collapse
Affiliation(s)
| | | | - José I. Borrell
- Grup de Química Farmacèutica, IQS School of Engineering, Universitat Ramon Llull, Via Augusta 390, E-08017 Barcelona, Spain; (J.D.); (R.P.d.l.B.)
| |
Collapse
|
3
|
Yun S, Nam G, Koo J. HiMolformer: Integrating graph and sequence representations for predicting liver microsome stability with SMILES. Comput Biol Chem 2024; 113:108263. [PMID: 39536405 DOI: 10.1016/j.compbiolchem.2024.108263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 09/13/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
In the initial stages of drug discovery or pre-clinical studies, understanding the metabolic stability of new molecules is crucial. Recently, research on pre-trained deep learning for molecular property prediction has been actively progressing, with various models being made open-source. However, most of these models rely on either 2D graph or 1D sequence for training, and the representation varies depending on the data format used. Consequently, combining multiple representations can broaden the scope of learning and may potentially be a manageable and most effective method to enhance performance. Therefore, we propose a novel hybrid model for predicting metabolic stability, which integrates representations from both graph-based and sequence-based models pre-trained for molecular features. This approach utilizes the combined strengths of 2D topological and 1D sequential information of molecules. HiMol, a graph-based graph neural network (GNN) model, and Molformer, a sequence-based Transformer model, were selected for integration, thus we named it HiMolformer. HiMolformer demonstrated superior performance compared to other models. We also focus on regression task for prediction with a empirical dataset from Korea Chemical Bank (KCB), comprising 3,498 molecules with mouse liver microsome (MLM) and human liver microsome (HLM) data obtained from actual metabolic reaction experiments. To the best of our knowledge, it is the first attempt to develop MLM and HLM prediction models using regression with a single SMILES input. The source code of this model is available at https://github.com/YUNSEOKWOO/HiMolformer.
Collapse
Affiliation(s)
- Seokwoo Yun
- Graduate School of Information and Communications, Sungkyunkwan University, Seoul, Republic of Korea.
| | - Gibeom Nam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Jahwan Koo
- Graduate School of Information and Communications, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Rehman B, Ahmed A, Khan S, Saleem N, Naseer F, Ahmad S. Exploring plant-based dengue therapeutics: from laboratory to clinic. Trop Dis Travel Med Vaccines 2024; 10:23. [PMID: 39543749 PMCID: PMC11566162 DOI: 10.1186/s40794-024-00232-1] [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: 07/04/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024] Open
Abstract
Dengue virus (DENV) is a mosquito-borne virus that causes dengue fever, a significant public health concern in many tropical and subtropical regions. Dengue is endemic in more than 100 countries, primarily in tropical and subtropical regions of the world. Each year, up to 400 million people get infected with dengue. Approximately 100 million people get sick from infection, and 40,000 die from severe dengue. Unfortunately, dengue vaccine development is also marred with various complicating factors, as the forefront candidate vaccine performed unsatisfactorily. Moreover, the only licensed vaccine (Dengvaxia) for children 9 through 16 years of age is available in just a few countries. The treatment difficulties are compounded by the absence of an effective antiviral agent. Exploring plant-based therapeutics for dengue from the laboratory to clinical application involves a multi-stage process, encompassing various scientific disciplines. Individual investigators have screened a wide range of plant extracts or compounds for potential antiviral activity against DENV. In vitro studies help identify candidates that exhibit inhibitory effects on viral replication. Some of the most promising medicinal plants showing in vitro activity against DENV include Andrographis paniculate, Acorus calamus, and Cladogynos orientalis. Further laboratory studies, both in vitro and in animal models (in vivo), elucidate the mechanisms of action by which the identified compounds exert antiviral effects. Medicinal plants such as Carica papaya, Cissampelos pareira, and Ipomea batata exhibited potent platelet-enhancing activities while Azadirachta indica and Curcuma longa showed promising effects in both in vitro and in vivo studies. Based on positive preclinical results, researchers design clinical trials. This involves careful planning of trial phases, patient recruitment criteria, ethical considerations, and endpoints. The most important medicinal plants showing efficacy and safety in clinical trials include Carica papaya and Cissampelos pareira. This review suggests that several promising medicinal plants exist that have the potential to be turned into clinical drugs to treat dengue infection. However, in addition to developing synthetic and plant-based therapies against dengue infection, vector management strategies should be made robust, emphasizing the need to focus on reducing disease incidence.
Collapse
Affiliation(s)
- Bisma Rehman
- Shifa College of Pharmaceutical Sciences, Shifa Tameer E Millat University, Islamabad, Pakistan
| | - Akhlaq Ahmed
- Department of Pharmacy, Hazara University, Mansehra, Pakistan
| | - Saeed Khan
- Dow International Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Nida Saleem
- Shifa College of Pharmaceutical Sciences, Shifa Tameer E Millat University, Islamabad, Pakistan
| | - Faiza Naseer
- Shifa College of Pharmaceutical Sciences, Shifa Tameer E Millat University, Islamabad, Pakistan.
- Department of Bioscience, Shifa Tameer E Millat University, Islamabad, Pakistan.
| | - Sagheer Ahmad
- Shifa College of Pharmaceutical Sciences, Shifa Tameer E Millat University, Islamabad, Pakistan.
| |
Collapse
|
5
|
Sadava D, Chen S. Molecular Interactions of the Plant Steroid Hormone Epibrassinolide on Human Drug-Sensitive and Drug-Resistant Small-Cell Lung Carcinoma Cells. Cancers (Basel) 2024; 16:3812. [PMID: 39594767 PMCID: PMC11592510 DOI: 10.3390/cancers16223812] [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: 10/22/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Small-cell lung cancer (SCLC) has a poor prognosis because it is often diagnosed after it has spread and develops multi-drug resistance. Epibrassinolide (EB) is a plant steroid hormone with widespread distribution and physiological effects. In plants, EB-activated gene expression occurs via a GSK-mediated signaling pathway, similar to Wnt-β-catenin signaling in animal cells that is elevated in cancer cells. Methods: This mechanistic parallel prompted investigations of the molecular interactions of EB on drug-sensitive (H69) and multi-drug-resistant (VPA) SCLC cells. Cellular and molecular investigations were performed. Results: Pharmacologic interactions between EB and the Wnt signaling inhibitors IGC-011 and PRI-724 were determined by the combination index method and showed antagonism, indicating that EB acts on the same pathway as these inhibitors. Following incubation of drug-sensitive and drug-resistant SCLC cells with EB, there was a reduction in β-catenin (e.g., 3.8 to 0.7 pg/µg protein), accompanied by a reduction in β-catenin promoter activity, measured by firefly luciferase-coupled promoter element transfection. Cellular β-catenin concentration is regulated by the active form of GSK3β. In Wnt signaling, active GSK3β is converted to inactive pGSK3β, thereby increasing the concentration of β-catenin. After incubation of SCLC cells with EB, there was a reduction in the inactive form (pGSK3β) and a relative increase in the active form (GSK3β). In vitro enzyme assays showed that EB did not inhibit purified GSK3β, but there was non-competitive inhibition when SCLC cell extracts were used as the source of enzyme. This indirect inhibition by EB indicates that it may act on the Wnt pathway by blocking the phosphorylation of GSK3β. The protein levels of three SCLC tumor markers, namely, NSE, CAV1, and MYCL1, were elevated in drug-resistant SCLC cells. EB incubation led to a significant reduction in the levels of the three markers. Two major effects of EB on SCLC cells are the promotion of apoptosis and the reversal of drug resistance. Transcriptional analyses showed that after exposure of SCLC cells to EB, there were increases in the expression of genes encoding apoptotic inducers (e.g., BAX and FAS) and effectors (e.g., CASP3) and reductions in the expression of genes encoding apoptosis inhibitors (e.g., survivin). PGP1 and MRP1, two membrane efflux pumps expressed in SCLC cells, were elevated in drug-resistant cells, but EB incubation did not affect these protein levels. Cellular assays of drug efflux by PGP1 showed an increase in drug-resistant cells, but EB did not alter efflux activity. Following exposure to human liver microsomes, EB was metabolized by NADPH-dependent oxidation and UDPG-dependent glucuronidation, as evidenced by the elimination of EB cytotoxicity against SCLC cells. Conclusions: Taken together, these data indicate that EB, a steroid hormone in plants consumed in the human diet, is pharmacologically active in drug-sensitive and drug-resistant SCLC cells in the Wnt signaling pathway, alters apoptotic gene expression, and is a substrate for microsomal modifications.
Collapse
Affiliation(s)
- David Sadava
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, 1500 E. Duarte Rd., Duarte, CA 91010, USA
| | | |
Collapse
|
6
|
Bošković J, Dobričić V, Savić J, Rupar J, Aleksić M, Marković B, Čudina O. In Vitro Evaluation of Pharmacokinetic Properties of Selected Dual COX-2 and 5-LOX Inhibitors. Pharmaceuticals (Basel) 2024; 17:1329. [PMID: 39458971 PMCID: PMC11510591 DOI: 10.3390/ph17101329] [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: 09/04/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Evaluation of pharmacokinetic properties is a significant step at the early stages of drug development. In this study, an in vitro evaluation of the pharmacokinetic properties of five newly synthesized compounds was performed. These compounds belong to N-hydroxyurea and hydroxamic acid derivatives and analogs of NSAIDs indomethacin, flurbiprofen, diclofenac, ibuprofen, and naproxen (compounds 1, 2, 3, 11, and 12, respectively) with dual COX-2 and 5-LOX inhibitory activity. Two in vitro methods (biopartitioning micellar chromatography (BMC) and PAMPA) were used to evaluate passive gastrointestinal absorption, while high-performance affinity chromatography (HPAC) and differential pulse voltammetry (DPV) were used to evaluate binding to human serum albumin (HSA). The introduction of N-hydroxyurea and hydroxamic acid groups into the structure of NSAIDs decreases both expected passive gastrointestinal absorption (BMC k values were from 3.02 to 9.50, while for NSAIDs were from 5.29 to 13.36; PAMPA -logPe values were between 3.81 and 4.76, while for NSAIDs were ≤3.46) and HSA binding (HPAC logk values were from 2.03 to 9.54, while for NSAIDs were ≥11.03; DPV peak potential shifts were between 7 and 34, while for NSAIDs were ≥54). Structural modifications of all tested compounds that increase lipophilicity could be considered to enhance their passive gastrointestinal absorption. Considering lower expected HSA binding and higher lipophilicity of tested compounds compared to corresponding NSAIDs, it can be expected that the volume of distribution of compounds 1, 2, 3, 11, and 12 will be higher. Reduced HSA binding may also decrease interactions with other drugs in comparison to corresponding NSAIDs. All tested compounds showed significant microsomal instability (25.07-58.44% decrease in concentration) in comparison to indomethacin (14.47%) and diclofenac (20.99%).
Collapse
Affiliation(s)
- Jelena Bošković
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Jelena Savić
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Jelena Rupar
- Department of Physical Chemistry and Instrumental Methods, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mara Aleksić
- Department of Physical Chemistry and Instrumental Methods, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Bojan Marković
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Olivera Čudina
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| |
Collapse
|
7
|
Hu J, Xia H, Chen X, Xu X, Wu HL, Shen Y, Xu RA, Wu W. Effect of isavuconazole on the pharmacokinetics of sunitinib and its mechanism. BMC Cancer 2024; 24:1131. [PMID: 39261851 PMCID: PMC11389264 DOI: 10.1186/s12885-024-12904-4] [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: 04/09/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND Sunitinib, a newly developed multi-targeted tyrosine kinase inhibitor (TKI), has become a common therapeutic option for managing advanced renal cell carcinoma (RCC). Examining the mechanism underlying the interaction between sunitinib and isavuconazole was the aim of this effort. METHODS The concentrations of sunitinib and its primary metabolite, N-desethyl sunitinib, were analyzed and quantified using ultra performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS). Our study evaluated the potential interaction between isavuconazole and sunitinib using rat liver microsomes (RLM), human liver microsomes (HLM), and in vivo rat models. For the in vivo study, two groups (n = 5) of Sprague-Dawley (SD) rats were randomly allocated to receive sunitinib either with or without co-administration of isavuconazole. Additionally, the effects of isavuconazole on the metabolic stability of sunitinib and N-desethyl sunitinib were studied in RLM in vitro. RESULTS Our findings demonstrated that in RLM, isavuconazole exhibited a mixed non-competitive and competitive inhibition mechanism, with an IC50 (half maximal inhibitory concentration) value of 1.33 µM. Meanwhile, in HLM, isavuconazole demonstrated a competitive inhibition mechanism, with an IC50 of 5.30 µM. In vivo studies showed that the presence of isavuconazole significantly increased the pharmacokinetic characteristics of sunitinib, with the AUC(0→t), AUC(0→∞), and Tmax rising to approximately 211.38%, 203.92%, and 288.89%, respectively, in contrast to the control group (5 mg/kg sunitinib alone). The pharmacokinetic characteristics of the metabolite N-desethyl sunitinib in the presence of isavuconazole remained largely unchanged compared to the control group. Furthermore, in vitro metabolic stability experiments revealed that isavuconazole inhibited the metabolic processing of both sunitinib and N-desethyl sunitinib. CONCLUSIONS Isavuconazole had a major impact on sunitinib metabolism, providing fundamental information for the precise therapeutic administration of sunitinib.
Collapse
Affiliation(s)
- Jinyu Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hailun Xia
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaohai Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xinhao Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hua-Lu Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxin Shen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ren-Ai Xu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Wenzhi Wu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Borsos E, Varga E, Aichinger G, Marko D. Unraveling Interspecies Differences in the Phase I Hepatic Metabolism of Alternariol and Alternariol Monomethyl Ether: Closing Data Gaps for a Comprehensive Risk Assessment. Chem Res Toxicol 2024; 37:1356-1363. [PMID: 39028893 PMCID: PMC11337205 DOI: 10.1021/acs.chemrestox.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024]
Abstract
The Alternaria mycotoxins alternariol (AOH) and alternariol 9-O-monomethyl ether (AME) are pervasive food contaminants known to exert adverse effects in vitro, yet their toxicokinetics remain inadequately understood. Thus, this study endeavors to elucidate the qualitative and quantitative aspects of the phase I metabolism of AOH and AME. To pursue this goal, reduced nicotinamide adenine dinucleotide phosphate (NADPH)-fortified porcine, rat, and human liver microsomes were incubated for 0-10 min with AOH or AME within a concentration range of 1-100 and 1-50 μM, respectively. The decline in the parent toxin concentration was monitored via liquid chromatography coupled to tandem mass spectrometry, whereas coupling to high-resolution mass spectrometry provided insights into the composition of the arising metabolic mixture. The collected quantitative data allowed us to calculate the hepatic intrinsic clearance rates of AOH and AME, marking a notable contribution to the field. Moreover, we unveiled interspecies differences in the pattern and rate of the phase I metabolism of the investigated mycotoxins. The presented findings lay the groundwork for physiologically based toxicokinetic modeling aimed at estimating local concentrations of these mycotoxins in specific organs, enhancing our understanding of their mode of action and adverse health effects.
Collapse
Affiliation(s)
- Eszter Borsos
- Department
of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Doctoral
School in Chemistry, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| | - Elisabeth Varga
- Department
of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Unit
Food Hygiene and Technology, Centre for Food Science and Veterinary
Public Health, Clinical Department for Farm Animals and Food System
Science, University of Veterinary Medicine,
Vienna, Vienna 1210, Austria
| | - Georg Aichinger
- Department
of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
- Department
of Health Sciences and Technology, ETH Zürich, Zürich 8092, Switzerland
| | - Doris Marko
- Department
of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna 1090, Austria
| |
Collapse
|
9
|
Lohr T, Scheuplein NJ, Jenkins C, Norville I, Erk C, Stapf M, Kirchner L, Sarkar-Tyson M, Holzgrabe U. Identification of active main metabolites of anti-infective inhibitors of the macrophage infectivity potentiator protein by liquid chromatography using mass detection. Arch Pharm (Weinheim) 2024; 357:e2400032. [PMID: 38687906 DOI: 10.1002/ardp.202400032] [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: 01/11/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024]
Abstract
Due to increasing antibiotic resistance, the development of anti-infectives with new mechanisms of action is crucial. Virulence factors such as the "macrophage infectivity potentiator" (Mip) protein, which catalyzes the folding of proline-containing proteins by means of their cis-trans isomerase (PPIase) activity, have come into focus as a potential new target. Since the inhibition of Mip by small molecules has been shown to lead to reduced virulence and survival in vitro, especially of Gram-negative bacteria such as Burkholderia pseudomallei (Bp), Neisseria meningitidis (Nm), and Neisseria gonorrhoeae (Ng), or Coxiella burnetii (Cb), among many others, a library of Mip inhibitors was developed. As drug metabolism has a significant impact on the overall therapeutic outcome, this report describes the biotransformation of the most potent Mip inhibitors. Therefore, the anti-infectives were treated using human liver microsomes in vitro. Liquid chromatography with tandem mass spectrometry (LC/MS-MS) methods were applied to identify the metabolites and quantify the metabolic degradation of the hit compounds. Active metabolites, N-oxides, were found, leading to new opportunities for further drug development.
Collapse
Affiliation(s)
- Theresa Lohr
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | | | | | - Isobel Norville
- DSTL, Defence Science and Technology Laboratory, Salisbury, UK
| | - Christine Erk
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Maximilian Stapf
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Lukas Kirchner
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| | - Mitali Sarkar-Tyson
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, Australia
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Würzburg, Germany
| |
Collapse
|
10
|
Palmeira-Mello MV, Costa AR, de Oliveira LP, Blacque O, Gasser G, Batista AA. Exploring the potential of ruthenium(II)-phosphine-mercapto complexes as new anticancer agents. Dalton Trans 2024; 53:10947-10960. [PMID: 38895770 DOI: 10.1039/d4dt01191k] [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: 06/21/2024]
Abstract
The search for new metal-based anticancer drug candidates is a fundamental task in medicinal inorganic chemistry. In this work, we assessed the potential of two new Ru(II)-phosphine-mercapto complexes as potential anticancer agents. The complexes, with the formula [Ru(bipy)(dppen)(Lx)]PF6 [(1), HL1 = 2-mercapto-pyridine and (2), HL2 = 2-mercapto-pyrimidine, bipy = 2,2'-bipyridine, dppen = cis-1,2-bis(diphenylphosphino)-ethylene] were synthesized and characterized by nuclear magnetic resonance (NMR) [1H, 31P(1H), and 13C], high resolution mass spectrometry (HR-MS), cyclic voltammetry, infrared and UV-Vis spectroscopies. Complex 2 was obtained as a mixture of two isomers, 2a and 2b, respectively. The composition of these metal complexes was confirmed by elemental analysis and liquid chromatography-mass spectrometry (LC-MS). To obtain insights into their lipophilicity, their distribution coefficients between n-octanol/PBS were determined. Both complexes showed affinity mainly for the organic phase, presenting positive log P values. Also, their stability was confirmed over 48 h in different media (i.e., DMSO, PBS and cell culture medium) via HPLC, UV-Vis and 31P{1H} NMR spectroscopies. Since enzymes from the P-450 system play a crucial role in cellular detoxification and metabolism, the microsomal stability of 1, which was found to be the most interesting compound of this study, was investigated using human microsomes to verify its potential oxidation in the liver. The analyses by LC-MS and ESI-MS reveal three main metabolites, obtained by oxidation in the dppen and bipy moieties. Moreover, 1 was able to interact with human serum albumin (HSA). The cytotoxicity of the metal complexes was tested in different cancerous and non-cancerous cell lines. Complex 1 was found to be more selective than cisplatin against MDA-MB-231 breast cancer cells when compared to MCF-10A non-cancerous cells. In addition, complex 1 affects cell morphology and migration, and inhibits colony formation in MDA-MB-231 cells, making it a promising cytotoxic agent against breast cancer.
Collapse
Affiliation(s)
- Marcos V Palmeira-Mello
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Paris, France.
| | - Analu R Costa
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| | - Leticia P de Oliveira
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| | - Olivier Blacque
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Gilles Gasser
- Chimie ParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Paris, France.
| | - Alzir A Batista
- Departament of Chemistry, Universidade Federal de São Carlos, 13561-901, São Carlos, SP, Brazil.
| |
Collapse
|
11
|
Petruncio G, Lee KH, Girgis M, Shellnutt Z, Beaulac Z, Xiang J, Lee SH, Peng X, Burdick M, Noble SM, Shim YM, Paige M. Synthesis and Evaluation of diaryl ether modulators of the leukotriene A 4 hydrolase aminopeptidase activity. Eur J Med Chem 2024; 272:116459. [PMID: 38704942 DOI: 10.1016/j.ejmech.2024.116459] [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: 03/28/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Activation of the aminopeptidase (AP) activity of leukotriene A4 hydrolase (LTA4H) presents a potential therapeutic strategy for resolving chronic inflammation. Previously, ARM1 and derivatives were found to activate the AP activity using the alanine-p-nitroanilide (Ala-pNA) as a reporter group in an enzyme kinetics assay. As an extension of this previous work, novel ARM1 derivatives were synthesized using a palladium-catalyzed Ullmann coupling reaction and screened using the same assay. Analogue 5, an aminopyrazole (AMP) analogue of ARM1, was found to be a potent AP activator with an AC50 of 0.12 μM. An X-ray crystal structure of LTA4H in complex with AMP was refined at 2.7 Å. Despite its AP activity with Ala-pNA substrate, AMP did not affect hydrolysis of the previously proposed natural ligand of LTA4H, Pro-Gly-Pro (PGP). This result highlights a discrepancy between the hydrolysis of more conveniently monitored chromogenic synthetic peptides typically employed in assays and endogenous peptides. The epoxide hydrolase (EH) activity of AMP was measured in vivo and the compound significantly reduced leukotriene B4 (LTB4) levels in a murine bacterial pneumonia model. However, AMP did not enhance survival in the murine pneumonia model over a 14-day period. A liver microsome stability assay showed metabolic stability of AMP. The results suggested that accelerated Ala-pNA cleavage is not sufficient for predicting therapeutic potential, even when the full mechanism of activation is known.
Collapse
Affiliation(s)
- Greg Petruncio
- Department of Chemistry & Biochemistry, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States; Center for Molecular Engineering, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States.
| | - Kyung Hyeon Lee
- Department of Chemistry & Biochemistry, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States; Center for Molecular Engineering, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States; Bacterial Diseases Branch, Wound Infections Department, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD, 20910, United States
| | - Michael Girgis
- Center for Molecular Engineering, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States; Department of Bioengineering, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States
| | - Zachary Shellnutt
- Department of Chemistry & Biochemistry, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States
| | - Zach Beaulac
- Department of Chemistry & Biochemistry, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States
| | - Jiangdong Xiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Soo Hyeon Lee
- Bacterial Diseases Branch, Wound Infections Department, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD, 20910, United States
| | - Xuejun Peng
- Bruker Scientific LLC., 101 Daggett Drive, San Jose CA, 95134, United States
| | - Marie Burdick
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, P.O. Box 800546, Charlottesville, VA, 22908, United States
| | - Schroeder M Noble
- Bacterial Diseases Branch, Wound Infections Department, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring, MD, 20910, United States.
| | - Yun M Shim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, P.O. Box 800546, Charlottesville, VA, 22908, United States.
| | - Mikell Paige
- Department of Chemistry & Biochemistry, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States; Center for Molecular Engineering, George Mason University, 10920 George Mason Circle, Manassas, VA, 20110, United States.
| |
Collapse
|
12
|
Shah P, Padilha EC, Kato R, Siramshetty VB, Huang W, Xu X. Consideration of vendor-related differences in hepatic metabolic stability data to optimize early ADME screening in drug discovery. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:34-39. [PMID: 37573009 PMCID: PMC10840824 DOI: 10.1016/j.slasd.2023.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/21/2023] [Accepted: 08/09/2023] [Indexed: 08/14/2023]
Abstract
Hepatic metabolic stability is a crucial determinant of oral bioavailability and plasma concentrations of a compound, and its measurement is important in early drug discovery. Preliminary metabolic stability estimations are commonly performed in liver microsomal fractions. At the National Center for Advancing Translational Sciences, a single-point assay in rat liver microsomes (RLM) is employed for initial stability assessment (Tier I) and a multi-point detailed stability assay is employed as a Tier II assay for promising compounds. Although the in vitro and in vivo metabolic stability of compounds typically exhibit good correlation, conflicting results may arise in certain cases. While investigating one such instance, we serendipitously found vendor-related RLM differences in metabolic stability and metabolite formation, which had implications for in vitro and in vivo correlations. In this study, we highlight the importance of considering vendor differences in hepatic metabolic stability data and discuss strategies to avoid these pitfalls.
Collapse
Affiliation(s)
- Pranav Shah
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States.
| | - Elias C Padilha
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States
| | - Rintaro Kato
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States
| | - Vishal B Siramshetty
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States; Department of Safety Assessment, Genentech, Inc., South San Francisco, CA 94080, United States
| | - Wenwei Huang
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States
| | - Xin Xu
- National Center for Advancing Translational Sciences (NCATS), 9800 Medical Center Drive, Rockville, MD 20850, United States
| |
Collapse
|
13
|
Tobajas Y, Alemany-Fornés M, Samarra I, Romero-Giménez J, Tintoré M, del Pino A, Canela N, del Bas JM, Ortega-Olivé N, de Lecea C, Escoté X. Diamine Oxidase Interactions with Anti-Inflammatory and Anti-Migraine Medicines in the Treatment of Migraine. J Clin Med 2023; 12:7502. [PMID: 38068554 PMCID: PMC10707353 DOI: 10.3390/jcm12237502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/09/2023] [Accepted: 12/01/2023] [Indexed: 07/03/2024] Open
Abstract
Histamine intolerance arises when there is a disparity between the production of histamine and the body's ability to break it down. In the gastrointestinal tract, the primary enzyme responsible for metabolizing ingested histamine is diamine oxidase (DAO), and a shortage of this enzyme has been associated with some diseases related to the respiratory, cardiovascular, nervous, muscular, and digestive systems, in addition to migraines. The treatment of migraines typically revolves around the utilization of both anti-migraine and anti-inflammatory drugs, but their interaction with DAO is not thoroughly understood. In this study, we examined the impact of nonsteroidal anti-inflammatory drugs (NSAIDs) and anti-migraine medications on DAO activity through in vitro experiments. We also investigated their effects on the human intestinal cell line Caco-2, assessing changes in DAO expression (both at the mRNA and protein levels) as well as DAO activity. The tested drugs, including ibuprofen, acetylsalicylic acid, paracetamol, a combination of acetylsalicylic acid with paracetamol and caffeine, zolmitriptan, and sumatriptan, did not inhibit DAO activity or reduce their levels. However, naproxen reduced DAO protein levels in human enterocyte cultures while not affecting DAO activity. These results suggest that combining anti-inflammatory and anti-migraine drugs with DAO enzyme supplementation for migraine patients with DAO deficiency could be beneficial for healthcare professionals in their daily practice.
Collapse
Affiliation(s)
- Yaiza Tobajas
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain; (Y.T.); (J.R.-G.); (N.O.-O.)
| | - Marc Alemany-Fornés
- DR Healthcare-AB Biotek HNH, 08017 Barcelona, Spain; (M.A.-F.); (M.T.); (C.d.L.)
| | - Iris Samarra
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain; (I.S.); (A.d.P.); (N.C.)
| | - Jordi Romero-Giménez
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain; (Y.T.); (J.R.-G.); (N.O.-O.)
| | - Maria Tintoré
- DR Healthcare-AB Biotek HNH, 08017 Barcelona, Spain; (M.A.-F.); (M.T.); (C.d.L.)
| | - Antoni del Pino
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain; (I.S.); (A.d.P.); (N.C.)
| | - Núria Canela
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain; (I.S.); (A.d.P.); (N.C.)
| | - Josep M. del Bas
- EURECAT, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain;
| | - Nàdia Ortega-Olivé
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain; (Y.T.); (J.R.-G.); (N.O.-O.)
| | - Carlos de Lecea
- DR Healthcare-AB Biotek HNH, 08017 Barcelona, Spain; (M.A.-F.); (M.T.); (C.d.L.)
| | - Xavier Escoté
- EURECAT, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain; (Y.T.); (J.R.-G.); (N.O.-O.)
- Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, Campus Sescelades, 43007 Tarragona, Spain
| |
Collapse
|
14
|
Tang Y, Xu L, Zhao J, Xiang P, Yan H. Metabolism of dipentylone in zebrafish and human liver microsomes determined by liquid chromatography-high resolution mass spectrometry. J Pharm Biomed Anal 2023; 236:115710. [PMID: 37690187 DOI: 10.1016/j.jpba.2023.115710] [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: 06/28/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
The consumption of novel psychoactive substances (NPS) is exceedingly prevalent in society, as these substances are sold and distributed as "legal highs." One novel synthetic cathinone emerging in the market is 1-(1,3-benzodioxol-5-yl)-2-(dimethylamino) pentan-1-one (dipentylone). The goal of this work was to study the in vivo and in vitro metabolism of dipentylone in zebrafish and human liver microsomes (HLMs) by liquid chromatography-high resolution mass spectrometry (LC-HRMS). The zebrafish and HLM samples contained 14 dipentylone metabolites, specifically 12 phase Ⅰ metabolites and 2 phase Ⅱ metabolites. The main metabolic pathways included monohydroxylation (M1 and M2), N-dealkylation (M3), hydroxylation of the aromatic ring and dealkoxylation of M3 (M4), O-dealkylation (M5), N-dealkylation of M5 (M6), reduction of carboxide (M7), monohydroxylation of M5 (M8), dehydrogenation (M9), dealkoxylation (M10), N-dealkylation of M10 (M11), dealkoxylation of M9 (M12), glucuronidation of M5 (M13), and sulfation (M14). The monohydroxylated metabolite (M2) can be recommended as metabolic markers for dipentylone. This study is the first to identify a target compound for monitoring the abuse of dipentylone and to determine the essential chemical structure of the metabolites for further toxicological research.
Collapse
Affiliation(s)
- Yiling Tang
- Department of Forensic Toxicology, Academy of Forensic Science, Shanghai Key Laboratory of Forensic Medicine, Shanghai 200063, China; Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Linhao Xu
- Department of Forensic Toxicology, Academy of Forensic Science, Shanghai Key Laboratory of Forensic Medicine, Shanghai 200063, China; Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Junbo Zhao
- Department of Forensic Toxicology, Academy of Forensic Science, Shanghai Key Laboratory of Forensic Medicine, Shanghai 200063, China
| | - Ping Xiang
- Department of Forensic Toxicology, Academy of Forensic Science, Shanghai Key Laboratory of Forensic Medicine, Shanghai 200063, China
| | - Hui Yan
- Department of Forensic Toxicology, Academy of Forensic Science, Shanghai Key Laboratory of Forensic Medicine, Shanghai 200063, China.
| |
Collapse
|
15
|
Chhatrapati Bisen A, Nashik Sanap S, Agrawal S, Biswas A, Sankar Bhatta R. Chemical metabolite synthesis and profiling: Mimicking in vivo biotransformation reactions. Bioorg Chem 2023; 139:106722. [PMID: 37453238 DOI: 10.1016/j.bioorg.2023.106722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 07/18/2023]
Abstract
Biotransformation was previously viewed as merely the structural characterization of drug metabolites, and it was performed only when drug candidates entered clinical development. The synthesis of drug metabolites is crucial to the drug development process because it generates either pharmacologically active, inactive, or reactive molecules and hence their characterization and comprehensive pharmacological evaluation is necessary. The chemical metabolite synthesis is very challenging due to the complex structures of many drug molecules, presence of multiple stereocenters, poor reaction yields, and the formation of unwanted by-products. Drug metabolites and their chemical synthesis have immense significance in the drug discovery process. The chemical synthesis of metabolites facilitates on- or off-target pharmacological and toxicological evaluations at the easiest. In a broader view metabolite could be a target lead molecule for drug design, toxic reactive metabolites, pharmaceutical standards for bioanalytical methods, etc. Collectively these metabolite information dossiers will aid regulatory agencies such as the EMA and FDA in maintaining strict vigilance over drug manufacturers with regard to the safety of NCE's and their hidden metabolites. Herein, we are presenting a systematic compilation of chemical and biocatalytic strategies reported to date for pharmaceutical drug metabolite synthesis. This review report is very useful for the laboratory synthesis of new drug metabolites, and their preclinical biological evaluation could aid in the detection of early threats (alerts) in drug discovery, eliminate the toxicity profile, explore newer pharmacology, and delivering a promising and safe drug candidate to humankind.
Collapse
Affiliation(s)
- Amol Chhatrapati Bisen
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sachin Nashik Sanap
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Sristi Agrawal
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Arpon Biswas
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Rabi Sankar Bhatta
- Pharmaceutics & Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
16
|
Keem MJ, Seo SW, Kim T, Jo BG, Kim SN, Yoon IS, Yang MH. A High-Performance Liquid Chromatography with Photodiode Array Detection Method for Simultaneous Determination of Three Compounds Isolated from Wikstroemia ganpi: Assessment of the Effects on Cytochrome P450-Mediated Metabolism In Vitro and In Vivo. Nutrients 2023; 15:4061. [PMID: 37764844 PMCID: PMC10534693 DOI: 10.3390/nu15184061] [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: 08/23/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
In natural products, the content and quality of the marker components differ depending on the part, production area, collection period, and extraction method; therefore, a standardized analysis method is required to obtain consistent results. This study developed a simultaneous analysis method for three marker components (7-methoxylutolin-5-O-glucoseide, pilloin 5-O-β-d-glucopyranoside, rutarensin) isolated and purified from Wikstroemia ganpi (W. ganpi). Simultaneous analysis was performed using high-performance liquid chromatography with photodiode array detection (HPLC-PDA) method that was validated according to the International Council for Harmonisation (ICH) guidelines. The developed analytical method exhibited linearity (r2 > 0.999), detection limits (0.72-3.34 μg/mL), and quantification limits (2.19-10.22 μg/mL). The relative standard deviation (RSD) value of intra- and inter-day precisions was less than 1.68%, and analyte recoveries (93.42-117.55%; RSD < 1.86%) were validated according to the analytical procedures, and all parameters were within the allowable range. Quantitative analysis of the three marker components from W. ganpi MeOH extract (WGM) showed 7-methoxylutolin-5-O-glucoseide with the highest content (51.81 mg/g). The inhibitory effects of WGM on cytochrome P450 (CYP) substrate drugs were further investigated. The in vitro study revealed that WGM inhibited the CYP3A-mediated metabolism of buspirone and that 7-methoxylutolin-5-O-glucoseide and pilloin 5-O-β-d-glucopyranoside inhibited the metabolism of buspirone with IC50 values of 2.73 and 18.7 μM, respectively. However, a single oral dose of WGM did not have significant effects on the pharmacokinetics of buspirone in rats, suggesting that WGM cannot function as an inhibitor of CYP3A-mediated metabolism in vivo.
Collapse
Affiliation(s)
- Min-Ji Keem
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (M.-J.K.); (B.-G.J.)
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
| | - Seong-Wook Seo
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
- Department of Manufacturing Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Taeyoung Kim
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
- Department of Manufacturing Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Beom-Geun Jo
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (M.-J.K.); (B.-G.J.)
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
| | - Su-Nam Kim
- Natural Products Research Institute, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea;
| | - In-Soo Yoon
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
- Department of Manufacturing Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min Hye Yang
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea; (M.-J.K.); (B.-G.J.)
- Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (S.-W.S.); (T.K.)
| |
Collapse
|
17
|
Tobajas Y, Alemany-Fornés M, Samarra I, Romero-Giménez J, Tintoré M, Del Pino A, Canela N, Del Bas JM, Ortega-Olivé N, de Lecea C, Escoté X. Interaction of Diamine Oxidase with Psychostimulant Drugs for ADHD Management. J Clin Med 2023; 12:4666. [PMID: 37510782 PMCID: PMC10380856 DOI: 10.3390/jcm12144666] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Histamine intolerance occurs when there is an imbalance between histamine production and the capacity for histamine degradation. Diamine oxidase (DAO) is the main enzyme for the catabolism of ingested histamine degradation in the gastrointestinal tract and its deficiency has been linked to allergy-like symptoms. Psychostimulant drugs are commonly used to treat Attention Deficit Hyperactivity Disorder (ADHD), but their interaction with DAO is not well characterized. In this work, we evaluated the effects of psychostimulant drugs (methylphenidate and lisdexamfetamine) on in vitro DAO activity and in the human cell line of enterocytes (Caco-2), evaluating DAO expression (mRNA and protein) and DAO activity. Methylphenidate and lisdexamfetamine did not repress the in vitro DAO activity. In addition, in Caco-2 cells, lisdexamfetamine promoted a strong upregulation of DAO mRNA levels, whereas methylphenidate tended to induce DAO activity. To sum up, methylphenidate and lisdexamfetamine treatments do not reduce DAO activity. These findings could be useful for physicians prescribing these two drugs to ADHD patients affected by DAO deficiency.
Collapse
Affiliation(s)
- Yaiza Tobajas
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain
| | | | - Iris Samarra
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain
| | - Jordi Romero-Giménez
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain
| | | | - Antoni Del Pino
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain
| | - Núria Canela
- Centre for Omic Sciences (COS), Joint Unit URV-EURECAT, Unique Scientific and Technical Infrastructures (ICTS), Eurecat, Centre Tecnològic de Catalunya, 43204 Reus, Spain
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| | - Nàdia Ortega-Olivé
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain
| | | | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health, 43204 Reus, Spain
| |
Collapse
|
18
|
Bassetto M, Zaluski J, Li B, Zhang J, Badiee M, Kiser PD, Tochtrop GP. Tuning the Metabolic Stability of Visual Cycle Modulators through Modification of an RPE65 Recognition Motif. J Med Chem 2023; 66:8140-8158. [PMID: 37279401 PMCID: PMC10824489 DOI: 10.1021/acs.jmedchem.3c00461] [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] [Indexed: 06/08/2023]
Abstract
In the eye, the isomerization of all-trans-retinal to 11-cis-retinal is accomplished by a metabolic pathway termed the visual cycle that is critical for vision. RPE65 is the essential trans-cis isomerase of this pathway. Emixustat, a retinoid-mimetic RPE65 inhibitor, was developed as a therapeutic visual cycle modulator and used for the treatment of retinopathies. However, pharmacokinetic liabilities limit its further development including: (1) metabolic deamination of the γ-amino-α-aryl alcohol, which mediates targeted RPE65 inhibition, and (2) unwanted long-lasting RPE65 inhibition. We sought to address these issues by more broadly defining the structure-activity relationships of the RPE65 recognition motif via the synthesis of a family of novel derivatives, which were tested in vitro and in vivo for RPE65 inhibition. We identified a potent secondary amine derivative with resistance to deamination and preserved RPE65 inhibitory activity. Our data provide insights into activity-preserving modifications of the emixustat molecule that can be employed to tune its pharmacological properties.
Collapse
Affiliation(s)
- Marco Bassetto
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Research Service, VA Long Beach Healthcare System, Long Beach, California 90822, United States
| | - Jordan Zaluski
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Bowen Li
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Jianye Zhang
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
| | - Mohsen Badiee
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Philip D Kiser
- Department of Physiology and Biophysics, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Ophthalmology, Gavin Herbert Eye Institute, Center for Translational Vision Research, School of Medicine, University of California - Irvine, Irvine, California 92697, United States
- Department of Clinical Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, University of California - Irvine, Irvine, California 92697, United States
- Research Service, VA Long Beach Healthcare System, Long Beach, California 90822, United States
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
19
|
Tran TMT, Addison RS, Davis RA, Rehm BHA. Bromotyrosine-Derived Metabolites from a Marine Sponge Inhibit Pseudomonas aeruginosa Biofilms. Int J Mol Sci 2023; 24:10204. [PMID: 37373352 DOI: 10.3390/ijms241210204] [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: 05/10/2023] [Revised: 06/06/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Pseudomonas aeruginosa forms stable biofilms, providing a major barrier for multiple classes of antibiotics and severely impairing treatment of infected patients. The biofilm matrix of this Gram-negative bacterium is primarily composed of three major exopolysaccharides: alginate, Psl, and Pel. Here, we studied the antibiofilm properties of sponge-derived natural products ianthelliformisamines A-C and their combinations with clinically used antibiotics. Wild-type P. aeruginosa strain and its isogenic exopolysaccharide-deficient mutants were employed to determine the interference of the compounds with biofilm matrix components. We identified that ianthelliformisamines A and B worked synergistically with ciprofloxacin to kill planktonic and biofilm cells. Ianthelliformisamines A and B reduced the minimum inhibitory concentration (MIC) of ciprofloxacin to 1/3 and 1/4 MICs, respectively. In contrast, ianthelliformisamine C (MIC = 53.1 µg/mL) alone exhibited bactericidal effects dose-dependently on both free-living and biofilm populations of wild-type PAO1, PAO1ΔpslA (Psl deficient), PDO300 (alginate overproducing and mimicking clinical isolates), and PDO300Δalg8 (alginate deficient). Interestingly, the biofilm of the clinically relevant mucoid variant PDO300 was more susceptible to ianthelliformisamine C than strains with impaired polysaccharide synthesis. Ianthelliformisamines exhibited low cytotoxicity towards HEK293 cells in the resazurin viability assay. Mechanism of action studies showed that ianthelliformisamine C inhibited the efflux pump of P. aeruginosa. Metabolic stability analyses indicated that ianthelliformisamine C is stable and ianthelliformisamines A and B are rapidly degraded. Overall, these findings suggest that the ianthelliformisamine chemotype could be a promising candidate for the treatment of P. aeruginosa biofilms.
Collapse
Affiliation(s)
- Tam M T Tran
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Russell S Addison
- Preclinical ADME/PK, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
| | - Rohan A Davis
- NatureBank, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia
| |
Collapse
|
20
|
Shahbazi Nia S, Hossain MA, Ji G, Jonnalagadda SK, Obeng S, Rahman MA, Sifat AE, Nozohouri S, Blackwell C, Patel D, Thompson J, Runyon S, Hiranita T, McCurdy CR, McMahon L, Abbruscato TJ, Trippier PC, Neugebauer V, German NA. Studies on diketopiperazine and dipeptide analogs as opioid receptor ligands. Eur J Med Chem 2023; 254:115309. [PMID: 37054561 PMCID: PMC10634475 DOI: 10.1016/j.ejmech.2023.115309] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
Using the structure of gliotoxin as a starting point, we have prepared two different chemotypes with selective affinity to the kappa opioid receptor (KOR). Using medicinal chemistry approaches and structure-activity relationship (SAR) studies, structural features required for the observed affinity were identified, and advanced molecules with favorable Multiparameter Optimization (MPO) and Ligand Lipophilicity (LLE) profiles were prepared. Using the Thermal Place Preference Test (TPPT), we have shown that compound2 blocks the antinociceptive effect of U50488, a known KOR agonist. Multiple reports suggest that modulation of KOR signaling is a promising therapeutic strategy in treating neuropathic pain (NP). As a proof-of-concept study, we tested compound 2 in a rat model of NP and recorded its ability to modulate sensory and emotional pain-related behaviors. Observed in vitro and in vivo results suggest that these ligands can be used to develop compounds with potential application as pain therapeutics.
Collapse
Affiliation(s)
- Siavash Shahbazi Nia
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Mohammad Anwar Hossain
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Sravan K Jonnalagadda
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Samuel Obeng
- Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University, Birmingham, AL, 35229, USA
| | - Md Ashrafur Rahman
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Ali Ehsan Sifat
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Collin Blackwell
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Dhavalkumar Patel
- Office of Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Jon Thompson
- Veterinary School of Medicine, Texas Tech University, Amarillo, TX, 79106, USA
| | - Scott Runyon
- Reserach Triangle Institute, Research Triangle Park, Durham, NC, 27709, USA
| | - Takato Hiranita
- Department of Pharmacology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Lance McMahon
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA; UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, 68106, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA; Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA
| | - Nadezhda A German
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA; Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
21
|
Li J, Choudhry N, Lv G, Nimishetti N, Reddy MC, Liu H, Allen TD, Zhang J, Yang D. In-vitro metabolism of LXY18, an orally available, potent blocker of AURKB relocation in mitosis. J Pharm Biomed Anal 2023; 232:115415. [PMID: 37120975 DOI: 10.1016/j.jpba.2023.115415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/02/2023] [Accepted: 04/21/2023] [Indexed: 05/02/2023]
Abstract
This study investigated the metabolism of LXY18, a quinolone-based compound that suppresses tumorigenesis by blocking AURKB localization. Metabolite profiling of LXY18 in liver microsomes from six species and human S9 fractions revealed that LXY18 undergoes various conserved metabolic reactions, such as N-hydroxylation, N-oxygenation, O-dealkylation, and hydrolysis, resulting in ten metabolites. These metabolites were produced through a combination of CYP450 enzymes, and non-CYP450 enzymes including CES1, and AO. Two metabolites, M1 and M2 were authenticated by chemically synthesized standards. M1 was the hydrolyzed product catalyzed by CES1 whereas M2 was a mono-N-oxidative derivative catalyzed by a CYP450 enzyme. AO was identified as the enzyme responsible for the formation of M3 with the help of AO-specific inhibitors and LXY18 analogs, 5b and 5c. M1 was the intermediate of LXY18 to produce M7, M8, M9, and M10. LXY18 potently inhibited 2C19 with an IC50 of 290 nM but had a negligible impact on the other CYP450s, indicating a low risk of drug-drug interaction. Altogether, the study provides valuable insights into the metabolic process of LXY18 and its suitability as a drug candidate. The data generated serves as a significant reference point for conducting further safety assessments and optimizing drug development.
Collapse
Affiliation(s)
- Jinhua Li
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| | - Namrta Choudhry
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| | - Gang Lv
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Naganna Nimishetti
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | | | - Hong Liu
- Anticancer Bioscience (US), South San Francisco, CA 94080, USA
| | | | - Jing Zhang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China
| | - Dun Yang
- Chengdu Anticancer Bioscience, Chengdu 610000, China; J. Michael Bishop Institute of Cancer Research, Chengdu 610000, China.
| |
Collapse
|
22
|
N H, C M, T R M, S S, S N, K E M, S C S, Y N, P V D, R N M. In Vitro Hepatic Models to Assess Herb-Drug Interactions: Approaches and Challenges. Pharmaceuticals (Basel) 2023; 16:ph16030409. [PMID: 36986508 PMCID: PMC10058280 DOI: 10.3390/ph16030409] [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: 02/06/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
A newfound appreciation for the benefits of herbal treatments has emerged in recent decades. However, herbal medication production still needs to establish standardized protocols that adhere to strict guidelines for quality assurance and risk minimization. Although the therapeutic effects of herbal medicines are extensive, the risk of herb-drug interactions remains a serious concern, limiting their use. Therefore, a robust, well-established liver model that can fully represent the liver tissue is required to study potential herb-drug interactions to ensure herbal medicines' safe and effective use. In light of this, this mini review investigates the existing in vitro liver models applicable to detecting herbal medicines' toxicity and other pharmacological targets. This article analyzes the benefits and drawbacks of existing in vitro liver cell models. To maintain relevance and effectively express the offered research, a systematic strategy was employed to search for and include all discussed studies. In brief, from 1985 to December 2022, the phrases "liver models", "herb-drug interaction", "herbal medicine", "cytochrome P450", "drug transporters pharmacokinetics", and "pharmacodynamics" were combined to search the electronic databases PubMed, ScienceDirect, and the Cochrane Library.
Collapse
Affiliation(s)
- Hlengwa N
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Masilela C
- Department of Biochemistry, North-West University, Mafikeng 2745, South Africa
| | - Mtambo T R
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
- Biomedical Research and Innovation Platform and South African Medical Research Council, Cape Town 7505, South Africa
| | - Sithole S
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
- Biomedical Research and Innovation Platform and South African Medical Research Council, Cape Town 7505, South Africa
| | - Naidoo S
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Machaba K E
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| | - Shabalala S C
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
- Biomedical Research and Innovation Platform and South African Medical Research Council, Cape Town 7505, South Africa
| | - Ntamo Y
- Biomedical Research and Innovation Platform and South African Medical Research Council, Cape Town 7505, South Africa
| | - Dludla P V
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
- Biomedical Research and Innovation Platform and South African Medical Research Council, Cape Town 7505, South Africa
| | - Milase R N
- Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3886, South Africa
| |
Collapse
|
23
|
Tupertsev B, Osipenko S, Kireev A, Nikolaev E, Kostyukevich Y. Simple In Vitro 18O Labeling for Improved Mass Spectrometry-Based Drug Metabolites Identification: Deep Drug Metabolism Study. Int J Mol Sci 2023; 24:ijms24054569. [PMID: 36902002 PMCID: PMC10002766 DOI: 10.3390/ijms24054569] [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: 02/06/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
The identification of drug metabolites formed with different in vitro systems by HPLC-MS is a standard step in preclinical research. In vitro systems allow modeling of real metabolic pathways of a drug candidate. Despite the emergence of various software and databases, identification of compounds is still a complex task. Measurement of the accurate mass, correlation of chromatographic retention times and fragmentation spectra are often insufficient for identification of compounds especially in the absence of reference materials. Metabolites can "slip under the nose", since it is often not possible to reliably confirm that a signal belongs to a metabolite and not to other compounds in complex systems. Isotope labeling has proved to be a tool that aids in small molecule identification. The introduction of heavy isotopes is done with isotope exchange reactions or with complicated synthetic schemes. Here, we present an approach based on the biocatalytic insertion of oxygen-18 isotope under the action of liver microsomes enzymes in the presence of 18O2. Using the local anesthetic bupivacaine as an example, more than 20 previously unknown metabolites were reliably discovered and annotated in the absence of the reference materials. In combination with high-resolution mass spectrometry and modern methods of mass spectrometric metabolism data processing, we demonstrated the ability of the proposed approach to increase the degree of confidence in interpretating metabolism data.
Collapse
Affiliation(s)
- Boris Tupertsev
- Center of Molecular and Cellular Biology (CMCB), Skolkovo Institute of Science and Technology, Nobel Str., 3, 121205 Moscow, Russia
- Moscow Institute of Physics and Technology, Phystech School of Biological and Medical Physics, Institutskiy per., 9, Dolgoprudny, 141701 Moscow, Russia
| | - Sergey Osipenko
- Center for Computational and Data-Intensive Science and Engineering, Skolkovo Institute of Science and Technology, Nobel Str., 3, 121205 Moscow, Russia
| | - Albert Kireev
- Center of Molecular and Cellular Biology (CMCB), Skolkovo Institute of Science and Technology, Nobel Str., 3, 121205 Moscow, Russia
| | - Eugene Nikolaev
- Center of Molecular and Cellular Biology (CMCB), Skolkovo Institute of Science and Technology, Nobel Str., 3, 121205 Moscow, Russia
| | - Yury Kostyukevich
- Center of Molecular and Cellular Biology (CMCB), Skolkovo Institute of Science and Technology, Nobel Str., 3, 121205 Moscow, Russia
- Correspondence:
| |
Collapse
|
24
|
Biotransformation of Penindolone, an Influenza A Virus Inhibitor. Molecules 2023; 28:molecules28031479. [PMID: 36771146 PMCID: PMC9920254 DOI: 10.3390/molecules28031479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
Penindolone (PND) is a novel broad-spectrum anti-Influenza A Virus (IAV) agent blocking hemagglutinin-mediated adsorption and membrane fusion. The goal of this work was to reveal the metabolic route of PND in rats. Ultra-high-performance liquid chromatography tandem high-resolution mass spectrometry (UHPLC-HRMS) was used for metabolite identification in rat bile, feces and urine after administration of PND. A total of 25 metabolites, including 9 phase I metabolites and 16 phase II metabolites, were characterized. The metabolic pathways were proposed, and metabolites were visualized via Global Natural Product Social Molecular Networking (GNPS). It was found that 65.24-80.44% of the PND presented in the formation of glucuronide conjugate products in bile, and more than 51% of prototype was excreted through feces. In in vitro metabolism of PND by rat, mouse and human liver microsomes (LMs) system, PND was discovered to be eliminated in LMs to different extents with significant species differences. The effects of chemical inhibitors of isozymes on the metabolism of PND in vitro indicated that CYP2E1/2C9/3A4 and UGT1A1/1A6/1A9 were the metabolic enzymes responsible for PND metabolism. PND metabolism in vivo could be blocked by UGTs inhibitor (ibrutinib) to a certain extent. These findings provided a basis for further research and development of PND.
Collapse
|
25
|
Roegner N, Pluym N, Peschel O, Leibold E, Kachhadia A, Scherer G, Scherer M. Determination of a specific metabolite for the non-ionic surfactant 2,4,7,9-Tetramethyl-5-decyne-4,7-diol (TMDD) by UPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2023; 1216:123584. [PMID: 36640715 DOI: 10.1016/j.jchromb.2022.123584] [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: 09/19/2022] [Revised: 11/09/2022] [Accepted: 12/23/2022] [Indexed: 12/31/2022]
Abstract
2,4,7,9-Tetramethyldec-5-yne-4,7-diol (TMDD) is a non-ionic surfactant commonly used as defoaming agent and numerous other applications. Effluents of wastewater treatment plants have been identified as one of the main sources of TMDD emissions into the environment. Due to its broad application in various fields, TMDD was selected for the development of a biomonitoring method for assessing human exposure within the frame of the cooperation project of the German Federal Ministry for the Environment, Nature Conservation, Building and Nuclear Safety (BMUB) and the German Chemical Industry Association (VCI) in 2020. This study aimed to identify a urinary metabolite for TMDD by UPLC-Q-Orbitrap-MS which can be used as a biomarker of TMDD exposure. Monohydroxylated TMDD (1-OH-TMDD) was deciphered as the most prominent metabolite of TMDD in humans in a series of in vitro and in vivo experiments. In a next step, a quantitative method for the determination of 1-OH-TMDD was developed and validated. Quantification was achieved by isotope dilution using D3-1-OH-TMDD as internal standard. The method is characterized by a simple sample clean-up procedure and an enzymatic hydrolysis of possible metabolite conjugates with ß-glucuronidase. Method validation was performed according to international guidelines for bioanalytical method validation. The method proved its robustness, precision, accuracy and sensitivity for the intended purpose, i.e. the assessment of TMDD exposure in the general population by means of human biomonitoring.
Collapse
Affiliation(s)
- Nadine Roegner
- Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Nikola Pluym
- Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Oliver Peschel
- Institut für Rechtsmedizin der Universität München, Nussbaumstr. 26, 80336 Munich, Germany
| | - Edgar Leibold
- BASF SE, Product Safety, 67056 Ludwigshafen, Germany
| | - Alpeshkumar Kachhadia
- Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Gerhard Scherer
- Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstr. 5, 82152 Planegg, Germany
| | - Max Scherer
- Analytisch-Biologisches Forschungslabor GmbH, Semmelweisstr. 5, 82152 Planegg, Germany.
| |
Collapse
|
26
|
Qualification of Human Liver Microsomes for Antibacterial Activity Screening of Drug Metabolites. Appl Microbiol 2023. [DOI: 10.3390/applmicrobiol3010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Microsomes are commonly used to perform in vitro drug metabolism, predominantly to form phase I drug metabolites. Pooled microsomes from multiple donors can contain microorganisms from underlying microbial diseases. Exposure to microbes can also occur during extraction if aseptic processing is compromised. Although microbial presence does not affect the metabolic activity of microsomes, presence of unwanted microorganisms can cause interference if the downstream application of microsomal drug metabolites is screening for antibacterial activity. In this work, traditional biochemical tests and advanced proteomics-based identification techniques were used to identify two gram-negative bacteria in pooled human liver microsomes. Several decontamination procedures were assessed to eradicate these two bacteria from the microsomes without affecting its metabolic capacity, and organic extraction was found to be the most convenient and efficient approach to decontaminate microsomes and screen drug metabolites for antibacterial activity against methicillin-resistant Staphylococcus aureus (MRSA).
Collapse
|
27
|
Balogh-Weiser D, Poppe L, Kenéz B, Decsi B, Koplányi G, Katona G, Gyarmati B, Ender F, Balogh GT. Novel biomimetic nanocomposite for investigation of drug metabolism. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Vargas E, Zhang F, Ben Hassine A, Ruiz-Valdepeñas Montiel V, Mundaca-Uribe R, Nandhakumar P, He P, Guo Z, Zhou Z, Fang RH, Gao W, Zhang L, Wang J. Using Cell Membranes as Recognition Layers to Construct Ultrasensitive and Selective Bioelectronic Affinity Sensors. J Am Chem Soc 2022; 144:17700-17708. [DOI: 10.1021/jacs.2c07956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Eva Vargas
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Fangyu Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Amira Ben Hassine
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Victor Ruiz-Valdepeñas Montiel
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Rodolfo Mundaca-Uribe
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Ponnusamy Nandhakumar
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Putian He
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Zhongyuan Guo
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Zhidong Zhou
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Ronnie H. Fang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Weiwei Gao
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Liangfang Zhang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| | - Joseph Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
29
|
Zhou J, Qian X, Zhou Y, Xiong S, Ji S, Wang Y, Zhao P. Human liver microsomes study on the inhibitory effect of plantainoside D on the activity of cytochrome P450 activity. BMC Complement Med Ther 2022; 22:197. [PMID: 35870998 PMCID: PMC9308932 DOI: 10.1186/s12906-022-03671-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Plantainoside D is widely existed in the herbs and possesses various pharmacological activities, making it possible to co-administrate with other herbs. Its effect on cytochrome P450 enzymes (P450) is a risk factor for inducing adverse drug-drug interactions. To assess the effect of plantainoside D on the activity of major P450 isoenzymes in human liver microsomes. METHODS The Cocktail method was conducted in human liver microsomes in the presence of probe substrates. The activity of P450 isoenzymes was evaluated by the production of corresponding metabolites. The concentration-dependent and time-dependent inhibition assays were performed in the presence of 0, 2.5, 5, 10, 25, 50, and 100 μM plantainoside D to characterize the inhibitory effect of plantainoside D. RESULTS Significant inhibition was observed in the activity of CYP1A2, 2D6, and 3A, which was concentration-dependent with the IC50 values of 12.83, 8.39, and 14.66 μM, respectively. The non-competitive manner and competitive manner were observed in the CYP3A inhibition (Ki = 7.16 μM) and CYP1A2 (Ki = 6.26 μM) and 2D6 inhibition (Ki = 4.54 μM), respectively. Additionally, the inhibition of CYP3A was found to be time-dependent with the KI of 1.28 μM-1 and Kinact of 0.039 min-1. CONCLUSIONS Weak inhibitory effects of plantainoside D on the activity of CYP1A2, 2D6, and 3A were revealed in vitro, implying its potential of inducing interactions with CYP1A2-, 2D6-, and 3A-metabolized drugs. Although further in vivo validations are needed, the feasibility of the Cocktail method in evaluating P450 activity has been verified.
Collapse
Affiliation(s)
- Jin Zhou
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Xian Qian
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Yanqing Zhou
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Shili Xiong
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, No.121 Luoxi Road, Baoshan District, Shanghai, 201908, China
| | - Shuxia Ji
- Department of Pharmacy, Shanghai Baoshan Luodian Hospital, Shanghai, 201908, China
| | - Ying Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Ping Zhao
- Clinical Research Center, Shanghai Baoshan Luodian Hospital, No.121 Luoxi Road, Baoshan District, Shanghai, 201908, China.
| |
Collapse
|
30
|
Landry ML, Trager R, Broccatelli F, Crawford JJ. When Cofactors Aren't X Factors: Functional Groups That Are Labile in Human Liver Microsomes in the Absence of NADPH. ACS Med Chem Lett 2022; 13:727-733. [PMID: 35450376 PMCID: PMC9014494 DOI: 10.1021/acsmedchemlett.2c00071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/09/2022] [Indexed: 11/28/2022] Open
Abstract
The metabolic stability of compounds is often assessed at an early stage in drug discovery programs by profiling with hepatic microsomes. Exclusion of the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH) in these assays provides insight into non-cytochrome P450 (CYP)-mediated metabolism. This report uses a matched molecular pair (MMP) application to assess which chemical substituents are commonly susceptible to non-NADPH-mediated metabolism by microsomes. The analysis found the overall prevalence of metabolism in the absence of NADPH to be low, with esters, amides, aldehydes, and oxetanes being among the most commonly susceptible functional groups. Given that non-CYP enzymes, such as esterases, may be expressed extrahepatically and lead to lower confidence in predicted pharmacokinetic profiles, an awareness of the functional groups that commonly undergo non-NADPH-mediated metabolism-as well as options for their replacement based on experimental MMP data-may help researchers derisk metabolic stability issues at an earlier stage in drug discovery.
Collapse
Affiliation(s)
- Matthew L. Landry
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard Trager
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Fabio Broccatelli
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - James J. Crawford
- Genentech, Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
31
|
Lee W, Il An G, Park H, Sarkar S, Ha YS, Huynh PT, Bhise A, Bhatt N, Ahn H, Pandya DN, Kim JY, Kim S, Jun E, Kim SC, Lee KC, Yoo J. Imaging Strategy that Achieves Ultrahigh Contrast by Utilizing Differential Esterase Activity in Organs: Application in Early Detection of Pancreatic Cancer. ACS NANO 2021; 15:17348-17360. [PMID: 34405675 DOI: 10.1021/acsnano.1c05165] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Most nanoparticles show much higher uptake in mononuclear phagocyte system (MPS) organs than in tumors, which has been a long-lasting dilemma in nanomedicine. Here, we report an imaging strategy that selectively decreases MPS organ uptakes by utilizing the differential esterase activity in tumors and other organs. When an esterase-labile radiotracer loaded liposome was injected into the body, radioactivity was rapidly excreted from the liver and spleen after breakage of the ester bond by esterase. However, the lipophilic radiotracer delivered to the tumor remained in the tumor with minimal bond cleavage. The underlying mechanism was fully characterized in vitro and in vivo in colon tumor models. As a proof of concept, the liposomal radiotracer was further optimized for the early detection of pancreatic cancer. The folate-coated liposomal radiotracer showed highly selective tumor uptake. At 4 h postinjection, a pancreatic tumor a few millimeters in size was unambiguously visualized in orthotopic tumor models by PET imaging. At 24 h, an exceptionally high tumor-to-background ratio was achieved, enabling the visualization of tumors alone with minimal background noise. More than 9% of the total radioactivity was found in the tumor. Utilizing our imaging strategy, various tumor imaging agents can be developed for sensitive detection with ultrahigh contrast.
Collapse
Affiliation(s)
- Woonghee Lee
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Gwang Il An
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Hyun Park
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Swarbhanu Sarkar
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Yeong Su Ha
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Phuong Tu Huynh
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Abhinav Bhise
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Nikunj Bhatt
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Heesu Ahn
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Darpan N Pandya
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jung Young Kim
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Seokho Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan 49315, Republic of Korea
| | - Eunsung Jun
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
- Department of Convergence Medicine, Asan Institute for Life Sciences, University of Ulsan College of Medicine and Asan Medical Center, Seoul 05505, Republic of Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, AMIST, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, Seoul 01812, Republic of Korea
| | - Jeongsoo Yoo
- Department of Molecular Medicine, Brain Korea 21 four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
32
|
Iqbal K, Pierce SH, Kozai K, Dhakal P, Scott RL, Roby KF, Vyhlidal CA, Soares MJ. Evaluation of Placentation and the Role of the Aryl Hydrocarbon Receptor Pathway in a Rat Model of Dioxin Exposure. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:117001. [PMID: 34747641 PMCID: PMC8574979 DOI: 10.1289/ehp9256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND Our environment is replete with chemicals that can affect embryonic and extraembryonic development. Dioxins, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are compounds affecting development through the aryl hydrocarbon receptor (AHR). OBJECTIVES The purpose of this investigation was to examine the effects of TCDD exposure on pregnancy and placentation and to evaluate roles for AHR and cytochrome P450 1A1 (CYP1A1) in TCDD action. METHODS Actions of TCDD were examined in wild-type and genome-edited rat models. Placenta phenotyping was assessed using morphological, biochemical, and molecular analyses. RESULTS TCDD exposures were shown to result in placental adaptations and at higher doses, pregnancy termination. Deep intrauterine endovascular trophoblast cell invasion was a prominent placentation site adaptation to TCDD. TCDD-mediated placental adaptations were dependent upon maternal AHR signaling but not upon placental or fetal AHR signaling nor the presence of a prominent AHR target, CYP1A1. At the placentation site, TCDD activated AHR signaling within endothelial cells but not trophoblast cells. Immune and trophoblast cell behaviors at the uterine-placental interface were guided by the actions of TCDD on endothelial cells. DISCUSSION We identified an AHR regulatory pathway in rats activated by dioxin affecting uterine and trophoblast cell dynamics and the formation of the hemochorial placenta. https://doi.org/10.1289/EHP9256.
Collapse
Affiliation(s)
- Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Stephen H. Pierce
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Regan L. Scott
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Katherine F. Roby
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Anatomy and Cell Biology, KUMC, Kansas City, Kansas, USA
| | - Carrie A. Vyhlidal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Obstetrics and Gynecology, KUMC, Kansas City, Kansas, USA
| |
Collapse
|
33
|
Rao Gajula SN, Pillai MS, Samanthula G, Sonti R. Cytochrome P450 enzymes: a review on drug metabolizing enzyme inhibition studies in drug discovery and development. Bioanalysis 2021; 13:1355-1378. [PMID: 34517735 DOI: 10.4155/bio-2021-0132] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Assessment of drug candidate's potential to inhibit cytochrome P450 (CYP) enzymes remains crucial in pharmaceutical drug discovery and development. Both direct and time-dependent inhibition of drug metabolizing CYP enzymes by the concomitant administered drug is the leading cause of drug-drug interactions (DDIs), resulting in the increased toxicity of the victim drug. In this context, pharmaceutical companies have grown increasingly diligent in limiting CYP inhibition liabilities of drug candidates in the early stages and examining risk assessments throughout the drug development process. This review discusses different strategies and decision-making processes for assessing the drug-drug interaction risks by enzyme inhibition and lays particular emphasis on in vitro study designs and interpretation of CYP inhibition data in a stage-appropriate context.
Collapse
Affiliation(s)
- Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Balanagar, Telangana, 50003, India
| | - Megha Sajakumar Pillai
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Balanagar, Telangana, 50003, India
| | - Gananadhamu Samanthula
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Balanagar, Telangana, 50003, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education & Research (NIPER), Hyderabad, Balanagar, Telangana, 50003, India
| |
Collapse
|
34
|
Sturm S, Högner C, Seger C, Stuppner H. Combining HPLC-DAD-QTOF-MS and HPLC-SPE-NMR to Monitor In Vitro Vitetrifolin D Phase I and II Metabolism. Metabolites 2021; 11:529. [PMID: 34436470 PMCID: PMC8400717 DOI: 10.3390/metabo11080529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
By combining HPLC-DAD-QTOF-MS and HPLC-SPE-NMR, the in vitro metabolism of vitetrifolin D, a pharmacologically active key molecule from Vitex agnus-castus in liver cell fractions, was investigated. Twenty-seven phase I and phase II metabolites were tentatively identified from the culture broth by HPLC-DAD-QTOF-MS. The subsequent HPLC-SPE-NMR analysis allowed for the unequivocal structural characterization of nine phase I metabolites. Since the preparative isolation of the metabolites was avoided, the substance input was much lower than in conventional strategies. The study did prove that the use of hyphenated instrumental analysis methodologies allows for the successful performance of in vitro metabolism studies, even if the availability of substances is very limited.
Collapse
|
35
|
Skibiński R, Trawiński J, Gawlik M. Characterization of Phase I Hepatic Metabolites of Anti-Premature Ejaculation Drug Dapoxetine by UHPLC-ESI-Q-TOF. Molecules 2021; 26:3794. [PMID: 34206424 PMCID: PMC8270242 DOI: 10.3390/molecules26133794] [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: 04/28/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 12/04/2022] Open
Abstract
Determination of the metabolism pathway of xenobiotics undergoing the hepatic pass is a crucial aspect in drug development since the presence of toxic biotransformation products may result in significant side effects during the therapy. In this study, the complete hepatic metabolism pathway of dapoxetine established according to the human liver microsome assay with the use of a high-resolution LC-MS system was described. Eleven biotransformation products of dapoxetine, including eight metabolites not reported in the literature so far, were detected and identified. N-dealkylation, hydroxylation, N-oxidation and dearylation were found to be the main metabolic reactions for the investigated xenobiotic. In silico analysis of toxicity revealed that the reaction of didesmethylation may contribute to the increased carcinogenic potential of dapoxetine metabolites. On the other hand, N-oxidation and aromatic hydroxylation biotransformation reactions possibly lead to the formation of mutagenic compounds.
Collapse
Affiliation(s)
- Robert Skibiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.T.); (M.G.)
| | | | | |
Collapse
|
36
|
Kaddah MMY, Talaat W, El Demellawy MA. Determination and structural characterization of ravidasvir metabolites by LC coupled to triple quadrupole linear ion trap MS: Application to pharmacokinetics and phase I metabolism in rats. Biomed Chromatogr 2021; 35:e5146. [PMID: 33893663 DOI: 10.1002/bmc.5146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023]
Abstract
Hepatitis C virus (HCV) is an infectious disease that has become a global clinical issue because of its significant morbidity and mortality. Novel anti-hepatitis C drugs are continuously developed to decrease the pervasiveness of the infection globally. A synthetic ravidasvir, benzimidazole-naphthylene-imidazole derivatives, has been used as an anti-HCV drug. This study determined the metabolites of ravidasvir and its pharmacokinetics in rats using information-dependent acquisition and multiple reaction monitoring scanning modes in linear ion trap LC-MS/MS instrument, respectively. Two time-programming linear-gradient chromatographic methods were employed using a Kinetex C18 column (50 × 3 mm, 2.6 μm) and a Luna HILIC column (100 × 4.6 mm, 3 μm) for the qualitative and quantitative determination of ravidasvir and its metabolites, respectively. In silico prediction where sites in a molecule are susceptible to metabolism by cytochrome P450 was implemented, which helped in proposing the metabolic pathway of ravidasvir. The most dominant metabolite in rat liver microsomal samples was oxidative ravidasvir, where one O-demethylated metabolite and eight isomers of the oxidative ravidasvir metabolites were identified. The study provides essential data for proposing the metabolic pathway and successfully applied it to determine the pharmacokinetics of ravidasvir in rat plasma.
Collapse
Affiliation(s)
- Mohamed Mohamed Yousri Kaddah
- Pharmaceutical and Fermentation Industries Development Center, City of Scientific Research and Technological Applications, New Borg El-Arab, Alexandria, Egypt
| | - Wael Talaat
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Maha A El Demellawy
- Center of Excellence for Preclinical Research in Drug Development, City of Scientific Research and Technological Applications, New Borg El-Arab, Alexandria, Egypt
| |
Collapse
|
37
|
Wegler C, Matsson P, Krogstad V, Urdzik J, Christensen H, Andersson TB, Artursson P. Influence of Proteome Profiles and Intracellular Drug Exposure on Differences in CYP Activity in Donor-Matched Human Liver Microsomes and Hepatocytes. Mol Pharm 2021; 18:1792-1805. [PMID: 33739838 PMCID: PMC8041379 DOI: 10.1021/acs.molpharmaceut.1c00053] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 01/07/2023]
Abstract
Human liver microsomes (HLM) and human hepatocytes (HH) are important in vitro systems for studies of intrinsic drug clearance (CLint) in the liver. However, the CLint values are often in disagreement for these two systems. Here, we investigated these differences in a side-by-side comparison of drug metabolism in HLM and HH prepared from 15 matched donors. Protein expression and intracellular unbound drug concentration (Kpuu) effects on the CLint were investigated for five prototypical probe substrates (bupropion-CYP2B6, diclofenac-CYP2C9, omeprazole-CYP2C19, bufuralol-CYP2D6, and midazolam-CYP3A4). The samples were donor-matched to compensate for inter-individual variability but still showed systematic differences in CLint. Global proteomics analysis outlined differences in HLM from HH and homogenates of human liver (HL), indicating variable enrichment of ER-localized cytochrome P450 (CYP) enzymes in the HLM preparation. This suggests that the HLM may not equally and accurately capture metabolic capacity for all CYPs. Scaling CLint with CYP amounts and Kpuu could only partly explain the discordance in absolute values of CLint for the five substrates. Nevertheless, scaling with CYP amounts improved the agreement in rank order for the majority of the substrates. Other factors, such as contribution of additional enzymes and variability in the proportions of active and inactive CYP enzymes in HLM and HH, may have to be considered to avoid the use of empirical scaling factors for prediction of drug metabolism.
Collapse
Affiliation(s)
- Christine Wegler
- Department
of Pharmacy, Uppsala University, 752 37 Uppsala, Sweden
- DMPK,
Research and Early Development Cardiovascular, Renal and Metabolism,
BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Pär Matsson
- Department
of Pharmacy, Uppsala University, 752 37 Uppsala, Sweden
| | - Veronica Krogstad
- Department
of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0315 Oslo, Norway
| | - Jozef Urdzik
- Department
of Surgical Sciences, Uppsala University, 751 85 Uppsala, Sweden
| | - Hege Christensen
- Department
of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, 0315 Oslo, Norway
| | - Tommy B. Andersson
- DMPK,
Research and Early Development Cardiovascular, Renal and Metabolism,
BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Per Artursson
- Department
of Pharmacy and Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
| |
Collapse
|
38
|
Parajuli P, Sable R, Shrestha L, Dahal A, Gauthier T, Taneja V, Jois S. Modulation of co-stimulatory signal from CD2-CD58 proteins by a grafted peptide. Chem Biol Drug Des 2021; 97:607-627. [PMID: 32946175 PMCID: PMC8717467 DOI: 10.1111/cbdd.13797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 06/23/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022]
Abstract
Peptides were designed to inhibit the protein-protein interaction of CD2 and CD58 to modulate the immune response. This work involved the design and synthesis of eight different peptides by replacing each amino acid residue in peptide 6 with alanine as well as grafting the peptide to the sunflower trypsin-inhibitor framework. From the alanine scanning studies, mutation at position 2 of the peptide was shown to result in increased potency to inhibit cell adhesion interactions. The most potent peptide from the alanine scanning was further studied for its detailed three-dimensional structure and binding to CD58 protein using surface plasmon resonance and flow cytometry. This peptide was used to graft to the sunflower trypsin inhibitor to improve the stability of the peptide. The grafted peptide, SFTI-a1, was further studied for its potency as well as its thermal, chemical, and enzymatic stability. The grafted peptide exhibited improved activity compared to our previously grafted peptide and was stable against thermal and enzymatic degradation.
Collapse
Affiliation(s)
- Pravin Parajuli
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201 USA
| | - Rushikesh Sable
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201 USA
| | - Leeza Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201 USA
| | - Achyut Dahal
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201 USA
| | - Ted Gauthier
- Biotechnology Laboratory, LSU AgCenter, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Veena Taneja
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Seetharama Jois
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe LA 71201 USA
| |
Collapse
|
39
|
Lange T, Thomas A, Görgens C, Bidlingmaier M, Schilbach K, Fichant E, Delahaut P, Thevis M. Comprehensive insights into the formation of metabolites of the ghrelin mimetics capromorelin, macimorelin and tabimorelin as potential markers for doping control purposes. Biomed Chromatogr 2021; 35:e5075. [PMID: 33458843 DOI: 10.1002/bmc.5075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/11/2021] [Accepted: 01/14/2021] [Indexed: 12/25/2022]
Abstract
Analytical methods to determine the potential misuse of the ghrelin mimetics capromorelin (CP-424,391), macimorelin (macrilen, EP-01572) and tabimorelin (NN703) in sports were developed. Therefore, different extraction strategies, i.e. solid-phase extraction, protein precipitation, as well as a "dilute-and-inject" approach, from urine and EDTA-plasma were assessed and comprehensive in vitro/in vivo experiments were conducted, enabling the identification of reliable target analytes by means of high resolution mass spectrometry. The drugs' biotransformation led to the preliminary identification of 51 metabolites of capromorelin, 12 metabolites of macimorelin and 13 metabolites of tabimorelin. Seven major metabolites detected in rat urine samples collected post-administration of 0.5-1.0 mg of a single oral dose underwent in-depth characterization, facilitating their implementation into future confirmatory test methods. In particular, two macimorelin metabolites exhibiting considerable abundances in post-administration rat urine samples were detected, which might contribute to an improved sensitivity, specificity, and detection window in case of human sports drug testing programs. Further, the intact drugs were implemented into World Anti-Doping Agency-compliant initial testing (limits of detection 0.02-0.60 ng/ml) and confirmation procedures (limits of identification 0.18-0.89 ng/ml) for human urine and blood matrices. The obtained results allow extension of the test spectrum of doping agents in multitarget screening assays for growth hormone-releasing factors from human urine.
Collapse
Affiliation(s)
- Tobias Lange
- Center for Preventive Doping Research/Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne, 50933, Germany
| | - Andreas Thomas
- Center for Preventive Doping Research/Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne, 50933, Germany
| | - Christian Görgens
- Center for Preventive Doping Research/Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne, 50933, Germany
| | - Martin Bidlingmaier
- Endocrine Laboratory, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstraße 1, Munich, 80336, Germany
| | - Katharina Schilbach
- Endocrine Laboratory, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ziemssenstraße 1, Munich, 80336, Germany
| | - Eric Fichant
- Département Santé, CER Groupe, Rue du Point du Jour 8, Marloie, 6900, Belgium
| | - Philippe Delahaut
- Département Santé, CER Groupe, Rue du Point du Jour 8, Marloie, 6900, Belgium
| | - Mario Thevis
- Center for Preventive Doping Research/Institute of Biochemistry, German Sport University Cologne, Am Sportpark Müngersdorf 6, Cologne, 50933, Germany.,European Monitoring Center for Emerging Doping Agents, Am Sportpark Müngersdorf 6, Cologne, 50933, Germany
| |
Collapse
|
40
|
Xiao L, Bei Y, Li J, Chen M, Zhang Y, Xiang Q. Preclinical Pharmacokinetics, Tissue Distribution and Primary Safety Evaluation of a Novel Curcumin Analogue H10 Suspension, a Potential 17β Hydroxysteroid Dehydrogenase Type 3 Inhibitor. Chem Pharm Bull (Tokyo) 2021; 69:52-58. [PMID: 33087639 DOI: 10.1248/cpb.c20-00242] [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] [Indexed: 11/22/2022]
Abstract
17β Hydroxysteroid dehydrogenase type 3 (17β-HSD3) is the key enzyme in the biosynthesis of testosterone, which is an attractive therapeutic target for prostate cancer (PCa). H10, a novel curcumin analogue, was identified as a potential 17β-HSD3 inhibitor. The pharmacokinetic study of H10 in rats were performed by intraperitoneal (i.p.), intravenous (i.v.) and oral (p.o.) administration. In addition, the inhibitory effects of H10 against liver CYP3A4 were investigated in vitro using human liver microsomes (HLMs). The acute and chronic toxicological characteristics were characterized using single-dose and 30 d administration. All the mice were alive after i.p. H10 with dose of no more than 100 mg/kg which are nearly the maximum solubility in acute toxicity test. The pharmacokinetic characteristics of H10 fitted with linear dynamics model after single dose. Furthermore, H10 could bioaccumulate in testis, which was the target organ of 17β-HSD3 inhibitor. H10 distributed highest in spleen, and then in liver both after single and multiple i.p. administration. Moreover, H10 showed weak inhibition towards liver CYP3A4, and did not cause significant changes in aspartate transaminase (AST) and alanine transaminase (ALT) levels after treated with H10 for continuously 30 d. Taken together, these preclinical characteristics laid the foundation for further clinical studies of H10.
Collapse
Affiliation(s)
- Lichun Xiao
- College of Pharmacy, Jinan University.,Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University
| | - Yu Bei
- Biopharmaceutical R&D Center of Jinan University
| | - Jian'an Li
- Biopharmaceutical R&D Center of Jinan University
| | - Minjie Chen
- Biopharmaceutical R&D Center of Jinan University
| | | | - Qi Xiang
- College of Pharmacy, Jinan University.,Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University.,Biopharmaceutical R&D Center of Jinan University
| |
Collapse
|
41
|
Assessment of Tissue Distribution and Metabolism of MP1, a Novel Pyrrolomycin, in Mice Using a Validated LC-MS/MS Method. Molecules 2020; 25:molecules25245898. [PMID: 33322110 PMCID: PMC7764159 DOI: 10.3390/molecules25245898] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 11/16/2022] Open
Abstract
MP1 is a novel marinopyrrole analogue with activity in MYCN amplified neuroblastoma cell lines. A rapid, selective, and sensitive liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) method was developed and validated for quantitation of MP1 in mouse plasma. Analyte separation was achieved using a Waters Acquity UPLC®BEH C18 column (1.7 µm, 100 × 2.1 mm). Mobile phase consisted of 0.1% acetic acid in water (10%) and methanol (90%) at a total flow rate of 0.25 mL/min. The mass spectrometer was operated at unit resolution in the multiple reaction monitoring (MRM) mode, using precursor ion > product ion transitions of 324.10 > 168.30 m/z for MP1 and 411.95 > 224.15 m/z for PL-3. The MS/MS response was linear over the concentration range from 0.2-500 ng/mL for MP1, correlation coefficient (r2) of 0.988. Precision (% RSD) and accuracy (% bias) were within the acceptable limits as per FDA guidelines. MP1 was stable under storage and laboratory handling conditions. The validated method was successfully applied to assess the solubility, in-vitro metabolism, plasma protein binding, and bio-distribution studies of MP1.
Collapse
|
42
|
Lei Y, Zhang B, Liu D, Zhao J, Dai X, Gao J, Mao Q, Feng Y, Zhao J, Lin F, Duan Y, Zhang Y, Bao Z, Yang Y, Mou Y, Wang S. Switching a Xanthine Oxidase Inhibitor to a Dual-Target Antagonist of P2Y1 and P2Y12 as an Oral Antiplatelet Agent with a Wider Therapeutic Window in Rats than Ticagrelor. J Med Chem 2020; 63:15752-15772. [DOI: 10.1021/acs.jmedchem.0c01524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Yu Lei
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Bing Zhang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Dan Liu
- Shenyang Hinewy Pharmaceutical Technology Co., Ltd., 41 Liutang Road, Shenhe District, Shenyang 110016, China
| | - Jian Zhao
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Xiwen Dai
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Jun Gao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Qing Mao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Yao Feng
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Jiaxing Zhao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Fengwei Lin
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Yulin Duan
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Yan Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Ziyang Bao
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Yuwei Yang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Yanhua Mou
- Department of Pharmacology, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| | - Shaojie Wang
- Key Laboratory of Structure-Based Drugs Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Culture Road, Shenhe District, Shenyang 110016, China
| |
Collapse
|
43
|
Magnetic Nanoparticles with Dual Surface Functions-Efficient Carriers for Metalloporphyrin-Catalyzed Drug Metabolite Synthesis in Batch and Continuous-Flow Reactors. NANOMATERIALS 2020; 10:nano10122329. [PMID: 33255480 PMCID: PMC7759782 DOI: 10.3390/nano10122329] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/14/2020] [Accepted: 11/18/2020] [Indexed: 01/02/2023]
Abstract
The dual functionalization of magnetic nanoparticles with inert (methyl) and reactive (aminopropyl) groups enables efficient immobilization of synthetic metalloporphyrins (such as 5,10,15,20-tetrakis(2,3,4,5,6-pentafluorophenyl)iron(II) porphyrin and 5,10,15,20-tetrakis-(4-sulfonatophenyl)iron(II) porphyrin) via covalent or ionic interactions. The proportion of reactive function on the surface has significant effect on the biomimetic activity of metalloporphyrins. The optimized magnetic nanocatalyst containing porphyrin was successfully applied for biomimetic oxidation of antihypertensive drug Amlodipine in batch and continuous-flow reactors as well.
Collapse
|
44
|
Ooka M, Lynch C, Xia M. Application of In Vitro Metabolism Activation in High-Throughput Screening. Int J Mol Sci 2020; 21:ijms21218182. [PMID: 33142951 PMCID: PMC7663506 DOI: 10.3390/ijms21218182] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023] Open
Abstract
In vitro methods which incorporate metabolic capability into the assays allow us to assess the activity of metabolites from their parent compounds. These methods can be applied into high-throughput screening (HTS) platforms, thereby increasing the speed to identify compounds that become active via the metabolism process. HTS was originally used in the pharmaceutical industry and now is also used in academic settings to evaluate biological activity and/or toxicity of chemicals. Although most chemicals are metabolized in our body, many HTS assays lack the capability to determine compound activity via metabolism. To overcome this problem, several in vitro metabolic methods have been applied to an HTS format. In this review, we describe in vitro metabolism methods and their application in HTS assays, as well as discuss the future perspectives of HTS with metabolic activity. Each in vitro metabolism method has advantages and disadvantages. For instance, the S9 mix has a full set of liver metabolic enzymes, but it displays high cytotoxicity in cell-based assays. In vitro metabolism requires liver fractions or the use of other metabolically capable systems, including primary hepatocytes or recombinant enzymes. Several newly developed in vitro metabolic methods, including HepaRG cells, three-dimensional (3D) cell models, and organ-on-a-chip technology, will also be discussed. These newly developed in vitro metabolism approaches offer significant progress in dissecting biological processes, developing drugs, and making toxicology studies quicker and more efficient.
Collapse
|
45
|
Wu Q, Tsuduki T. CYP4F13 is the Major Enzyme for Conversion of alpha-Eleostearic Acid into cis-9, trans-11-Conjugated Linoleic Acid in Mouse Hepatic Microsomes. J Oleo Sci 2020; 69:1061-1075. [PMID: 32879197 DOI: 10.5650/jos.ess20080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Our previous studies have shown that α-eleostearic acid (α-ESA; cis-9, trans-11, trans-13 (c9,t11,t13)-conjugated linolenic acid (CLnA)) is converted into c9,t11-conjugated linoleic acid (CLA) in rats. Furthermore, we have demonstrated that the conversion of α-ESA into CLA is a nicotinamide adenine dinucleotide phosphate (NADPH)-dependent enzymatic reaction, which occurs mostly in the rat liver. However, the precise metabolic pathway and enzyme involved have not been identified yet. Therefore, in this study we aimed to determine the role of cytochrome P450 (CYP) in the conversion of α-ESA into c9,t11-CLA using an in vitro reconstitution system containing mouse hepatic microsomes, NADPH, and α-ESA. The CYP4 inhibitors, 17-ODYA and HET0016, performed the highest level of inhibition of CLA formation. Furthermore, the redox partner cytochrome P450 reductase (CPR) inhibitor, 2-chloroethyl ethyl sulfide (CEES), also demonstrated a high level of inhibition. Thus, these results indicate that the NADPH-dependent CPR/CYP4 system is responsible for CLA formation. In a correlation analysis between the specific activity of CLA formation and Cyp4 family gene expression in tissues, Cyp4a14 and Cyp4f13 demonstrated the best correlations. However, the CYP4F substrate prostaglandin A1 (PGA1) exhibited the strongest inhibitory effect on CLA formation, while the CYP4A and CYP4B1 substrate lauric acid had no inhibitory effect. Therefore, we conclude that the CYP4F13 enzyme is the major enzyme involved in CLA formation. This pathway is a novel pathway for endogenous CLA synthesis, and this study provides insight into the potential application of CLnA in functional foods.
Collapse
Affiliation(s)
- Qiming Wu
- Laboratory of Food and Biomolecular Science, Graduate School of Agriculture, Tohoku University
| | - Tsuyoshi Tsuduki
- Laboratory of Food and Biomolecular Science, Graduate School of Agriculture, Tohoku University
| |
Collapse
|
46
|
Understanding performance of 3D-printed sorbent in study of metabolic stability. J Chromatogr A 2020; 1629:461501. [PMID: 32841768 DOI: 10.1016/j.chroma.2020.461501] [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: 03/02/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 11/21/2022]
Abstract
Metabolic stability tests are one of the fundamental steps at the preclinical stages of new drug development. Microsomes, used as a typical enzymatic model of liver biotransformation, can be a challenging matrix for analytical scientists due to a high concentration of cellular proteins and membrane lipids. In the work, we propose a new procedure integrating biotransformation reaction with SPME-like protocol for sample clean-up. It is beneficial to increase the overall quality of results in contrary to the typical protein precipitation approach. A set of ten arylpiperazine analogs, six of which are considered promising drug candidates (and four are accepted drugs) were used as a probe to assess the goodness of the newly proposed approach. In order to promote an efficient extraction protocol, a new, miniaturized shape of a sorbent, suitable to perform the extraction in 100 µL of the sample has been designed. Termination of the biotransformation process by protein denaturation with hot water was additionally evaluated. A quantitative structure-property relationship (QSPR) study using Orthogonal Partial Least Squares (OPLS) technique to reveal insights to the sorption mechanism was also performed. The obtained results showed the new 3D-printed sorbent can be an attractive basis for the new sample preparation approach for metabolic stability studies and an alternative for commercially available protocols based on solid-phase microextraction (SPME) or solid-phase extraction (SPE) principles.
Collapse
|
47
|
Miners JO, Rowland A, Novak JJ, Lapham K, Goosen TC. Evidence-based strategies for the characterisation of human drug and chemical glucuronidation in vitro and UDP-glucuronosyltransferase reaction phenotyping. Pharmacol Ther 2020; 218:107689. [PMID: 32980440 DOI: 10.1016/j.pharmthera.2020.107689] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/26/2022]
Abstract
Enzymes of the UDP-glucuronosyltransferase (UGT) superfamily contribute to the elimination of drugs from almost all therapeutic classes. Awareness of the importance of glucuronidation as a drug clearance mechanism along with increased knowledge of the enzymology of drug and chemical metabolism has stimulated interest in the development and application of approaches for the characterisation of human drug glucuronidation in vitro, in particular reaction phenotyping (the fractional contribution of the individual UGT enzymes responsible for the glucuronidation of a given drug), assessment of metabolic stability, and UGT enzyme inhibition by drugs and other xenobiotics. In turn, this has permitted the implementation of in vitro - in vivo extrapolation approaches for the prediction of drug metabolic clearance, intestinal availability, and drug-drug interaction liability, all of which are of considerable importance in pre-clinical drug development. Indeed, regulatory agencies (FDA and EMA) require UGT reaction phenotyping for new chemical entities if glucuronidation accounts for ≥25% of total metabolism. In vitro studies are most commonly performed with recombinant UGT enzymes and human liver microsomes (HLM) as the enzyme sources. Despite the widespread use of in vitro approaches for the characterisation of drug and chemical glucuronidation by HLM and recombinant enzymes, evidence-based guidelines relating to experimental approaches are lacking. Here we present evidence-based strategies for the characterisation of drug and chemical glucuronidation in vitro, and for UGT reaction phenotyping. We anticipate that the strategies will inform practice, encourage development of standardised experimental procedures where feasible, and guide ongoing research in the field.
Collapse
Affiliation(s)
- John O Miners
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Andrew Rowland
- Department of Clinical Pharmacology and Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | | | | | | |
Collapse
|
48
|
Noonepalle S, Shen S, Ptáček J, Tavares MT, Zhang G, Stránský J, Pavlíček J, Ferreira GM, Hadley M, Pelaez G, Bařinka C, Kozikowski AP, Villagra A. Rational Design of Suprastat: A Novel Selective Histone Deacetylase 6 Inhibitor with the Ability to Potentiate Immunotherapy in Melanoma Models. J Med Chem 2020; 63:10246-10262. [PMID: 32815366 DOI: 10.1021/acs.jmedchem.0c00567] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Selective inhibition of histone deacetylase 6 (HDAC6) is being recognized as a therapeutic approach for cancers. In this study, we designed a new HDAC6 inhibitor, named Suprastat, using in silico simulations. X-ray crystallography and molecular dynamics simulations provide strong evidence to support the notion that the aminomethyl and hydroxyl groups in the capping group of Suprastat establish significant hydrogen bond interactions, either direct or water-mediated, with residues D460, N530, and S531, which play a vital role in regulating the deacetylase function of the enzyme and which are absent in other isoforms. In vitro characterization of Suprastat demonstrates subnanomolar HDAC6 inhibitory potency and a hundred- to a thousand-fold HDAC6 selectivity over the other HDAC isoforms. In vivo studies reveal that a combination of Suprastat and anti-PD1 immunotherapy enhances antitumor immune response, mediated by a decrease of protumoral M2 macrophages and increased infiltration of antitumor CD8+ effector and memory T-cells.
Collapse
Affiliation(s)
- Satish Noonepalle
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Sida Shen
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jakub Ptáček
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Maurício T Tavares
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Guiping Zhang
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Jan Stránský
- Centre of Molecular Structure, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Jiří Pavlíček
- Centre of Molecular Structure, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | - Glaucio M Ferreira
- Department of Pharmacy, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP 05508-000, Brazil
| | - Melissa Hadley
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Guido Pelaez
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| | - Cyril Bařinka
- Laboratory of Structural Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, Vestec 252 50, Czech Republic
| | | | - Alejandro Villagra
- Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, District of Columbia 20052, United States
| |
Collapse
|
49
|
Heck CS, Seneviratne HK, Bumpus NN. Twelfth-Position Deuteration of Nevirapine Reduces 12-Hydroxy-Nevirapine Formation and Nevirapine-Induced Hepatocyte Death. J Med Chem 2020; 63:6561-6574. [PMID: 32065749 PMCID: PMC7959450 DOI: 10.1021/acs.jmedchem.9b01990] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Indexed: 01/08/2023]
Abstract
Cytochrome P450-dependent metabolism of the anti-HIV drug nevirapine (NVP) to 12-hydroxy-NVP (12-OHNVP) has been implicated in NVP toxicities. We investigated the impact of twelfth-position trideuteration (12-D3NVP) on the hepatic metabolism of and response to NVP. Formation of 12-OHNVP decreased in human (10.6-fold) and mouse (4.6-fold) hepatocytes incubated with 10 μM 12-D3NVP vs NVP. An observed kinetic isotope effect of 10.1 was measured in human liver microsomes. During mouse hepatocyte treatment (400 μM) with NVP or 12-D3NVP, cell death was reduced 30% with 12-D3NVP vs NVP, while glucuronidated and glutathione-conjugated metabolites increased with 12-D3NVP vs NVP. Using mass spectrometry proteomics, changes in hepatocyte protein expression, including an increase in stress marker insulin-like growth factor-binding protein 1 (IGFBP-1), were observed with 12-D3NVP vs NVP. These results demonstrate that while deuteration can reduce P450 metabolite formation, impacts on phase II metabolism and hepatocyte protein expression should be considered when employing deuteration to reduce P450 metabolite-related hepatotoxicity.
Collapse
Affiliation(s)
- Carley
J. S. Heck
- Department
of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Herana Kamal Seneviratne
- Department
of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Namandjé N. Bumpus
- Department
of Pharmacology & Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Medicine, Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
50
|
Renn A, Su B, Liu H, Sun J, Tseng YJ. Advances in the prediction of mouse liver microsomal studies: From machine learning to deep learning. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alex Renn
- Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei City Taiwan
- Department of Molecular and Cellular Biology University of California‐Berkeley Berkeley California USA
| | - Bo‐Han Su
- Department of Computer Science and Information Engineering National Taiwan University Taipei City Taiwan
| | - Hsin Liu
- Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei City Taiwan
| | - Joseph Sun
- Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei City Taiwan
| | - Yufeng J. Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics National Taiwan University Taipei City Taiwan
- Department of Computer Science and Information Engineering National Taiwan University Taipei City Taiwan
| |
Collapse
|