1
|
Steinbauer FF, Lehr T, Reichel A. Exploring the Potential of Adaptive, Local Machine Learning in Comparison to the Prediction Performance of Global Models: A Case Study from Bayer's Caco-2 Permeability Database. J Chem Inf Model 2024; 64:9163-9172. [PMID: 39564926 DOI: 10.1021/acs.jcim.4c01083] [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: 11/21/2024]
Abstract
Machine learning (ML) techniques are being widely implemented to fill the gap in simple molecular design guidelines for newer therapeutic modalities in the extended and beyond rule of five chemical space (eRo5, bRo5). These ML techniques predict molecular properties directly from the structure, allowing for the prioritization of promising compounds. However, the performance of models varies greatly among ML use cases. A molecular property for which achieving sufficient performance in generalizing global models still remains difficult is Caco-2 permeability. Especially within the lower permeability ranges, which are specific for larger molecules belonging to the e/bRo5 space, accurate regression predictions have proven to be challenging. The present study, therefore, identifies a suitable combination of ML algorithm and descriptors, consisting of the LightGBM algorithm and RDKit molecular property descriptors, to predict Caco-2 permeability very efficiently by a simple global model. An additionally introduced local model uses the same algorithm and descriptors but selects its training data based on Tanimoto fingerprint similarity to match the individual test compound's structure. Evaluation of this adaptive model, by systematically varying the number of most similar structures for training, shows that, in comparison to the global model, there was only marginally improved performance with specific training data constellations. These random improvements indicate that deriving general rules for local model parametrization is not possible a priori for the chosen algorithm and descriptor combination, and preselecting training data does not seem advantageous over global ML based on all available data, while creation of more data-efficient models was generally proven to be possible.
Collapse
Affiliation(s)
- Frank Filip Steinbauer
- Preclinical Modeling and Simulation, Preclinical Development, Bayer AG, Muellerstr. 178, 13353 Berlin, Germany
- Department of Clinical Pharmacy, Saarland University, 66123 Saarbruecken, Germany
| | - Thorsten Lehr
- Department of Clinical Pharmacy, Saarland University, 66123 Saarbruecken, Germany
| | - Andreas Reichel
- Preclinical Modeling and Simulation, Preclinical Development, Bayer AG, Muellerstr. 178, 13353 Berlin, Germany
| |
Collapse
|
2
|
Alberto-Silva AS, Hemmer S, Bock HA, da Silva LA, Scott KR, Kastner N, Bhatt M, Niello M, Jäntsch K, Kudlacek O, Bossi E, Stockner T, Meyer MR, McCorvy JD, Brandt SD, Kavanagh P, Sitte HH. Bioisosteric analogs of MDMA: Improving the pharmacological profile? J Neurochem 2024; 168:2022-2042. [PMID: 38898705 PMCID: PMC11449655 DOI: 10.1111/jnc.16149] [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: 11/15/2023] [Revised: 03/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, 'ecstasy') is re-emerging in clinical settings as a candidate for the treatment of specific neuropsychiatric disorders (e.g. post-traumatic stress disorder) in combination with psychotherapy. MDMA is a psychoactive drug, typically regarded as an empathogen or entactogen, which leads to transporter-mediated monoamine release. Despite its therapeutic potential, MDMA can induce dose-, individual-, and context-dependent untoward effects outside safe settings. In this study, we investigated whether three new methylenedioxy bioisosteres of MDMA improve its off-target profile. In vitro methods included radiotracer assays, transporter electrophysiology, bioluminescence resonance energy transfer and fluorescence-based assays, pooled human liver microsome/S9 fraction incubations, metabolic stability studies, isozyme mapping, and liquid chromatography coupled to high-resolution mass spectrometry. In silico methods included molecular docking. Compared with MDMA, all three MDMA bioisosteres (ODMA, TDMA, and SeDMA) showed similar pharmacological activity at human serotonin, dopamine, and norepinephrine transporters (hSERT, hDAT, and hNET, respectively) but decreased agonist activity at 5-HT2A/2B/2C receptors. Regarding their hepatic metabolism, they differed from MDMA, with N-demethylation being the only metabolic route shared, and without forming phase II metabolites. In addition, TDMA showed an enhanced intrinsic clearance in comparison to its congeners. Additional screening for their interaction with human organic cation transporters (hOCTs) and plasma membrane monoamine transporter (hPMAT) revealed a weaker interaction of the MDMA analogs with hOCT1, hOCT2, and hPMAT. Our findings suggest that these new MDMA bioisosteres might constitute appealing therapeutic alternatives to MDMA, sparing the primary pharmacological activity at hSERT, hDAT, and hNET, but displaying a reduced activity at 5-HT2A/2B/2C receptors and alternative hepatic metabolism. Whether these MDMA bioisosteres may pose lower risk alternatives to the clinically re-emerging MDMA warrants further studies.
Collapse
Affiliation(s)
- Ana Sofia Alberto-Silva
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Selina Hemmer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Hailey A. Bock
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Leticia Alves da Silva
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Kenneth R. Scott
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St James Hospital, Dublin, Ireland
| | - Nina Kastner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Manan Bhatt
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Marco Niello
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Kathrin Jäntsch
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Oliver Kudlacek
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Elena Bossi
- Laboratory of Cellular and Molecular Physiology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
- Center for Research in Neuroscience, University of Insubria, Varese, Italy
| | - Thomas Stockner
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus R. Meyer
- Department of Experimental and Clinical Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg, Germany
| | - John D. McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Simon D. Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, United Kingdom
| | - Pierce Kavanagh
- Department of Pharmacology and Therapeutics, School of Medicine, Trinity Centre for Health Sciences, St James Hospital, Dublin, Ireland
| | - Harald H. Sitte
- Center for Physiology and Pharmacology, Institute of Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, 19328 Amman, Jordan
- Center for Addiction Research and Science, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
3
|
Meng D, Ren S. Mairin from Huangqi Decoction Mitigates Liver Cirrhosis through
Suppression of Pro-inflammatory Signaling Pathways: A Network
Pharmacology and Experimental Study. THE NATURAL PRODUCTS JOURNAL 2024; 14. [DOI: 10.2174/0122103155273345231210170121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/25/2023] [Accepted: 11/03/2023] [Indexed: 01/04/2025]
Abstract
Background::
Liver cirrhosis is a consequence of various chronic liver conditions and may
lead to liver failure and cancer. Huangqi Decoction (HQD) is a Traditional Chinese Medicine (TCM)
effective for treating liver conditions, including cirrhosis. Therefore, both the active ingredients and
the pharmacological actions of HQD deserve further exploration. The active components and pharmacological
actions of HQD in preventing and treating liver cirrhosis were investigated using network
pharmacology. The actions of the principal active ingredient, Mairin, were investigated empirically.
Methods::
Using network pharmacology, the critical components of HQD were identified from multiple
databases, and UPLC screening and targets were investigated using Swiss Target Prediction.
Targets associated with liver cirrhosis were identified using the GeneCards database. GO and KEGG
enrichment analysis of targets that overlapped between HQD and cirrhosis were analyzed in DAVID,
and a “component-target-pathway” network for HQD was created in Cytoscape 3.7.2. The biological
functions of the key active component, Mairin, were investigated using in silico docking, cell experiments,
and evaluation in a carbon-tetrachloride (CCl4)-induced mouse model of liver cirrhosis.
CCK-8 and F-actin assays were used to measure cell viability and hepatic stellate cell (HSC) activation,
respectively; fibrosis was measured by histological and immunohistochemical evaluations, and
the levels of the cirrhosis-related protein α-SMA and predicted essential target proteins in the PI3KAKT,
NFκB-IκBα, and NLRP3-IL18 pathways were determined by western blotting.
Results::
Fourteen active HQD components, 72 targets, and 10 pathways common to HQD and cirrhosis
were identified. Network analysis indicated the association of Mairin with most targets and
with inflammation through the PI3K/Akt, NF-κB, and NLRP3 pathways. Dose-dependent reductions
in the activation and proliferation of LX-2 cells after Mairin treatment were observed. Mairin reversed
the histopathological changes in the livers of cirrhosis model mice. Mairin also significantly
reduced the α-SMA, NF-κB, IκBα, NLRP3, and IL-18 protein levels while increasing those of p-
PI3K and p-Akt, suggesting that Mairin mitigates liver cirrhosis through modulation of the PI3KAKT,
NFκB-IκBα, and NLRP3-IL18 pathways.
Conclusions::
Using a comprehensive investigative process involving network pharmacology, bioinformatics,
and experimental verification, it was found that Mairin, an active component of HQD,
may be useful for developing specific treatments for preventing and treating liver cirrhosis.
Collapse
Affiliation(s)
- Di Meng
- Department of Traditional Chinese Medicine, The First Hospital of China Medical University, Shenyang, China
| | - Shuang Ren
- Department of Traditional Chinese Medicine, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Dong L, Zhuang X. Insights into Inhalation Drug Disposition: The Roles of Pulmonary Drug-Metabolizing Enzymes and Transporters. Int J Mol Sci 2024; 25:4671. [PMID: 38731891 PMCID: PMC11083391 DOI: 10.3390/ijms25094671] [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: 03/17/2024] [Revised: 04/14/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
The past five decades have witnessed remarkable advancements in the field of inhaled medicines targeting the lungs for respiratory disease treatment. As a non-invasive drug delivery route, inhalation therapy offers numerous benefits to respiratory patients, including rapid and targeted exposure at specific sites, quick onset of action, bypassing first-pass metabolism, and beyond. Understanding the characteristics of pulmonary drug transporters and metabolizing enzymes is crucial for comprehending efficient drug exposure and clearance processes within the lungs. These processes are intricately linked to both local and systemic pharmacokinetics and pharmacodynamics of drugs. This review aims to provide a comprehensive overview of the literature on lung transporters and metabolizing enzymes while exploring their roles in exogenous and endogenous substance disposition. Additionally, we identify and discuss the principal challenges in this area of research, providing a foundation for future investigations aimed at optimizing inhaled drug administration. Moving forward, it is imperative that future research endeavors to focus on refining and validating in vitro and ex vivo models to more accurately mimic the human respiratory system. Such advancements will enhance our understanding of drug processing in different pathological states and facilitate the discovery of novel approaches for investigating lung-specific drug transporters and metabolizing enzymes. This deeper insight will be crucial in developing more effective and targeted therapies for respiratory diseases, ultimately leading to improved patient outcomes.
Collapse
Affiliation(s)
| | - Xiaomei Zhuang
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China;
| |
Collapse
|
5
|
Alberto-Silva AS, Hemmer S, Bock HA, Alves da Silva L, Scott KR, Kastner N, Bhatt M, Niello M, Jäntsch K, Kudlacek O, Bossi E, Stockner T, Meyer MR, McCorvy JD, Brandt SD, Kavanagh P, Sitte HH. Bioisosteric analogs of MDMA with improved pharmacological profile. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.08.588083. [PMID: 38645142 PMCID: PMC11030374 DOI: 10.1101/2024.04.08.588083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
3,4-Methylenedioxymethamphetamine (MDMA, ' ecstasy' ) is re-emerging in clinical settings as a candidate for the treatment of specific psychiatric disorders (e.g. post-traumatic stress disorder) in combination with psychotherapy. MDMA is a psychoactive drug, typically regarded as an empathogen or entactogen, which leads to transporter-mediated monoamine release. Despite its therapeutic potential, MDMA can induce dose-, individual-, and context-dependent untoward effects outside safe settings. In this study, we investigated whether three new methylenedioxy bioisosteres of MDMA improve its off-target profile. In vitro methods included radiotracer assays, transporter electrophysiology, bioluminescence resonance energy transfer and fluorescence-based assays, pooled human liver microsome/S9 fraction incubation with isozyme mapping, and liquid chromatography coupled to high-resolution mass spectrometry. In silico methods included molecular docking. Compared with MDMA, all three MDMA bioisosteres (ODMA, TDMA, and SeDMA) showed similar pharmacological activity at human serotonin and dopamine transporters (hSERT and hDAT, respectively) but decreased activity at 5-HT 2A/2B/2C receptors. Regarding their hepatic metabolism, they differed from MDMA, with N -demethylation being the only metabolic route shared, and without forming phase II metabolites. Additional screening for their interaction with human organic cation transporters (hOCTs) and plasma membrane transporter (hPMAT) revealed a weaker interaction of the MDMA analogs with hOCT1, hOCT2, and hPMAT. Our findings suggest that these new MDMA analogs might constitute appealing therapeutic alternatives to MDMA, sparing the primary pharmacological activity at hSERT and hDAT, but displaying a reduced activity at 5-HT 2A/2B/2C receptors and reduced hepatic metabolism. Whether these MDMA bioisosteres may pose lower risk alternatives to the clinically re-emerging MDMA warrants further studies.
Collapse
|
6
|
Li XQ, Elebring M, Dahlén A, Weidolf L. In Vivo Metabolite Profiles of an N-Acetylgalactosamine-Conjugated Antisense Oligonucleotide AZD8233 Using Liquid Chromatography High-Resolution Mass Spectrometry: A Cross-Species Comparison in Animals and Humans. Drug Metab Dispos 2023; 51:1350-1361. [PMID: 37429729 DOI: 10.1124/dmd.123.001370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/12/2023] Open
Abstract
AZD8233, a liver-targeting antisense oligonucleotide (ASO), inhibits subtilisin/kexin type 9 protein synthesis. It is a phosphorothioated 3-10-3 gapmer with a central DNA sequence flanked by constrained 2'-O-ethyl 2',4'-bridged nucleic acid (cEt-BNA) wings and conjugated to a triantennary N-acetylgalactosamine (GalNAc) ligand at the 5'-end. Herein we report the biotransformation of AZD8233, as given by liver, kidney, plasma and urine samples, after repeated subcutaneous administration to humans, mice, rats, rabbits, and monkeys. Metabolite profiles were characterized using liquid chromatography high-resolution mass spectrometry. Metabolite formation was consistent across species, mainly comprising hydrolysis of GalNAc sugars, phosphodiester-linker hydrolysis releasing the full-length ASO, and endonuclease-mediated hydrolysis within the central DNA gap followed by exonuclease-mediated 5'- or 3'-degradation. All metabolites contained the 5'- or 3'-cEt-BNA terminus. Most shortmer metabolites had the free terminal alcohol at 5'- and 3'-positions of ribose, although six were found retaining the terminal 5'-phosphorothioate group. GalNAc conjugated shortmer metabolites were also observed in urine. Synthesized metabolite standards were applied for (semi)quantitative metabolite assessment. Intact AZD8233 was the major component in plasma, whereas the unconjugated full-length ASO was predominant in tissues. In plasma, most metabolites were shortmers retaining the 3'-cEt-BNA terminus, whereas metabolites containing the 5'- or 3'-cEt-BNA terminus were detected in both tissues and urine. All metabolites in human plasma were also detected in all nonclinical species, and all human urine metabolites were detected in monkey urine. In general, metabolite profiles in animal species were qualitatively similar and quantitatively exceeded the exposures of the circulating metabolites in humans at the doses studied. SIGNIFICANCE STATEMENT: This study presents metabolite identification and profiling of AZD8233, an N-acetylgalactosamine-conjugated antisense oligonucleotide (ASO), across species. A biotransformation strategy for ASOs was established by utilizing biologic samples collected from toxicology and/or clinical studies and liquid chromatography high-resolution mass spectrometry analysis without conducting bespoke radiolabeled absorption, distribution, metabolism, and excretion studies. The generated biotransformation package was considered adequate by health authorities to progress AZD8233 into a phase 3 program, proving its applicability to future metabolism studies of ASOs in drug development.
Collapse
Affiliation(s)
- Xue-Qing Li
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (X.-Q.L., M.E., L.W.); and Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (A.D.)
| | - Marie Elebring
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (X.-Q.L., M.E., L.W.); and Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (A.D.)
| | - Anders Dahlén
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (X.-Q.L., M.E., L.W.); and Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (A.D.)
| | - Lars Weidolf
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (X.-Q.L., M.E., L.W.); and Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (A.D.)
| |
Collapse
|
7
|
Datta-Mannan A, Molitoris BA, Feng Y, Martinez MM, Sandoval RM, Brown RM, Merkel D, Croy JE, Dunn KW. Intravital Microscopy Reveals Unforeseen Biodistribution Within the Liver and Kidney Mechanistically Connected to the Clearance of a Bifunctional Antibody. Drug Metab Dispos 2023; 51:403-412. [PMID: 36460476 PMCID: PMC11022859 DOI: 10.1124/dmd.122.001049] [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: 08/03/2022] [Revised: 10/16/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Bifunctional antibody (BfAb) therapeutics offer the potential for novel functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including changes in pharmacokinetics that limit the compound's therapeutic profile. A better understanding of how molecular modifications affect in vivo tissue interactions could help inform BfAb design. The present studies were predicated on the observation that a BfAb designed to have minimal off-target interactions cleared from the circulation twice as fast as the monoclonal antibody (mAb) from which it was derived. The present study leverages the spatial and temporal resolution of intravital microscopy (IVM) to identify cellular interactions that may explain the different pharmacokinetics of the two compounds. Disposition studies of mice demonstrated that radiolabeled compounds distributed similarly over the first 24 hours, except that BfAb accumulated approximately two- to -three times more than mAb in the liver. IVM studies of mice demonstrated that both distributed to endosomes of liver endothelia but with different kinetics. Whereas mAb accumulated rapidly within the first hour of administration, BfAb accumulated only modestly during the first hour but continued to accumulate over 24 hours, ultimately reaching levels similar to those of the mAb. Although neither compound was freely filtered by the mouse or rat kidney, BfAb, but not mAb, was found to accumulate over 24 hours in endosomes of proximal tubule cells. These studies demonstrate how IVM can be used as a tool in drug design, revealing unpredicted cellular interactions that are undetectable by conventional analyses. SIGNIFICANCE STATEMENT: Bifunctional antibodies offer novel therapeutic functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including undesirable changes in pharmacokinetics. Studies of the dynamic distribution of a bifunctional antibody and its parent monoclonal antibody presented here demonstrate how intravital microscopy can expand our understanding of the in vivo disposition of therapeutics, detecting off-target interactions that could not be detected by conventional pharmacokinetics approaches or predicted by conventional physicochemical analyses.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Bruce A Molitoris
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Yiqing Feng
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Michelle M Martinez
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Ruben M Sandoval
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Robin M Brown
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Daniel Merkel
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Johnny E Croy
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Kenneth W Dunn
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| |
Collapse
|
8
|
Study on In Vitro Metabolism and In Vivo Pharmacokinetics of Beauvericin. Toxins (Basel) 2022; 14:toxins14070477. [PMID: 35878215 PMCID: PMC9320654 DOI: 10.3390/toxins14070477] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 01/25/2023] Open
Abstract
Beauvericin (BEA) is a well-known mycotoxin produced by many fungi, including Beaveria bassiana. The purpose of this study was to evaluate the in vitro distribution and metabolism characteristics as well as the in vivo pharmacokinetic (PK) profile of BEA. The in vitro metabolism studies of BEA were performed using rat, dog, mouse, monkey and human liver microsomes, cryopreserved hepatocytes and plasma under conditions of linear kinetics to estimate the respective elimination rates. Additionally, LC-UV-MSn (n = 1~2) was used to identify metabolites in human, rat, mouse, dog and monkey liver microsomes. Furthermore, cytochrome P450 (CYP) reaction phenotyping was carried out. Finally, the absolute bioavailability of BEA was evaluated by intravenous and oral administration in rats. BEA was metabolically stable in the liver microsomes and hepatocytes of humans and rats; however, it was a strong inhibitor of midazolam 1′-hydroxylase (CYP3A4) and mephenytoin 4′-hydroxylase (CYP2C19) activities in human liver microsomes. The protein binding fraction values of BEA were >90% and the half-life (T1/2) values of BEA were approximately 5 h in the plasma of the five species. The absolute bioavailability was calculated to be 29.5%. Altogether, these data indicate that BEA has great potential for further development as a drug candidate. Metabolic studies of different species can provide important reference values for further safety evaluation.
Collapse
|
9
|
Zhao G, Tong Y, Luan F, Zhu W, Zhan C, Qin T, An W, Zeng N. Alpinetin: A Review of Its Pharmacology and Pharmacokinetics. Front Pharmacol 2022; 13:814370. [PMID: 35185569 PMCID: PMC8854656 DOI: 10.3389/fphar.2022.814370] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022] Open
Abstract
Flavonoids isolated from medicinal herbs have been utilized as valuable health-care agents due to their virous biological applications. Alpinetin is a natural flavonoid that emerges in many widely used medicinal plants, and has been frequently applied in Chinese patent drugs. Accumulated evidence has demonstrated that alpinetin possesses a broad range of pharmacological activities such as antitumor, antiinflammation, hepatoprotective, cardiovascular protective, lung protective, antibacterial, antiviral, neuroprotective, and other properties through regulating multiple signaling pathways with low systemic toxicity. However, pharmacokinetic studies have documented that alpinetin may have poor oral bioavailability correlated to its extensive glucuronidation. Currently, the reported pharmacological properties and pharmacokinetics profiles of alpinetin are rare to be scientifically reviewed. In this article, we aimed to highlight the mechanisms of action of alpinetin in various diseases to strongly support its curative potentials for prospective clinical applications. We also summarized the pharmacokinetics properties and proposed some viable strategies to convey an appreciable reference for future advances of alpinetin in drug development.
Collapse
|
10
|
Zhao D, Feng SX, Zhang HJ, Zhang N, Liu XF, Wan Y, Zhou YX, Li JS. Pharmacokinetics, tissue distribution and excretion of five rhubarb anthraquinones in rats after oral administration of effective fraction of anthraquinones from rheum officinale. Xenobiotica 2021; 51:916-925. [PMID: 34110981 DOI: 10.1080/00498254.2021.1940353] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Rhubarb, a famous traditional Chinese medicine, shows a wide range of physiological activities and pharmacological benefits. Rhubarb anthraquinones are perceived as the pharmacologically active compounds of Rhubarb, and understanding metabolism of them is crucial to assure safety and effectiveness of clinical application. In this study, the pharmacokinetics, tissue distribution and excretion of five rhubarb anthraquinones (aloe-emodin, rhein, emodin, chrysophanol, physcion) were systematically investigated after oral administration of rhubarb extract to rats.An HPLC method was developed and validated for quantitation of five rhubarb anthraquinones in rat plasma, tissues, urine and faeces to investigate the Pharmacokinetic characteristics. The results showed that the proposed method was suitable for the quantification of five anthraquinones in plasma, tissue and excreta samples with satisfactory linear (r > 0.99), precision (<10%) and recovery (85.12-104.20%). The plasma concentration profiles showed a quick absorption with the mean Tmax of 0.42-0.75 h and t1/2 of 6.60-15.11 h for five anthraquinones. The analytes were widely distributed in most of the tissues. Approximately 0.13-10.59% and 28.47-81.14% of five anthraquinones were recovered in urine and faeces within 132 h post-dosing, which indicated the major elimination route was faeces excretion.In summary, this study lays a foundation for elucidating the pharmacokinetic rule of rhubarb anthraquinone and the important data can provide reliable scientific resource for further research.
Collapse
Affiliation(s)
- Di Zhao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, China
| | - Su-Xiang Feng
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, China.,Zhengzhou Key Laboratory of Chinese Medicine Quality Control and Evaluation, Zhengzhou, China
| | - Hao-Jie Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, China
| | - Na Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xue-Fang Liu
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, China
| | - Yan Wan
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Zhengzhou Key Laboratory of Chinese Medicine Quality Control and Evaluation, Zhengzhou, China
| | | | - Jian-Sheng Li
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, China.,Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases Co-Constructed by Henan Province & Education Ministry of P. R. China, Zhengzhou, China.,Zhengzhou Key Laboratory of Chinese Medicine Quality Control and Evaluation, Zhengzhou, China
| |
Collapse
|
11
|
A review of pharmacological and pharmacokinetic properties of Forsythiaside A. Pharmacol Res 2021; 169:105690. [PMID: 34029711 DOI: 10.1016/j.phrs.2021.105690] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
Traditional Chinese medicine plays a significant role in the treatment of various diseases and has attracted increasing attention for clinical applications. Forsythiae Fructus, the dried fruit of Forsythia suspensa (Thunb.) Vahl, is a widely used Chinese medicinal herb in clinic for its extensive pharmacological activities. Forsythiaside A is the main active index component isolated from Forsythiae Fructus and possesses prominent bioactivities. Modern pharmacological studies have confirmed that Forsythiaside A exhibits significant activities in treating various diseases, including inflammation, virus infection, neurodegeneration, oxidative stress, liver injury, and bacterial infection. In this review, the pharmacological activities of Forsythiaside A have been comprehensively reviewed and summarized. According to the data, Forsythiaside A shows remarkable anti-inflammation, antivirus, neuroprotection, antioxidant, hepatoprotection, and antibacterial activities through regulating multiple signaling transduction pathways such as NF-κB, MAPK, JAK/STAT, Nrf2, RLRs, TRAF, TLR7, and ER stress. In addition, the toxicity and pharmacokinetic properties of Forsythiaside A are also discussed in this review, thus providing a solid foundation and evidence for further studies to explore novel effective drugs from Chinese medicine monomers.
Collapse
|
12
|
Universal Automated Immunoaffinity Purification-CE-MS Platform for Accelerating Next Generation Biologic Design. Anal Chem 2021; 93:5562-5569. [PMID: 33764735 DOI: 10.1021/acs.analchem.1c00149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
As the pharmaceutical industry places greater emphasis on pairing biological pathways with appropriate therapeutic intervention, an increase in the use of biologic drugs has emerged. With increasing complexity of biotherapeutics, absorption, distribution, metabolism, and excretion (ADME) studies have also become increasingly complex. The characterization of ADME properties is critical to tuning the pharmacokinetic profiles of next generation biologics (NGBs). The knowledge of the fate of a drug is essential for the enhancement of the design processes, elongation of exposure at the desired site of action, and achieving efficacy with minimum toxicity. In vivo proteolytic cleavage of biotherapeutics may lead to undesirable in vivo properties, such as rapid clearance, low bioavailability, and loss of pharmacodynamic effect. All of these may affect drug efficacy and/or generate safety concerns through increases in immunogenicity or off-target toxicity. The work herein describes the development of a robust, fully automated immunoaffinity purification (IA)-capillary electrophoresis-mass spectrometry (CE-MS) workflow. The reagents were carefully optimized to maximize isolation yields while minimizing the number of experimental steps to analytical results. The result is the development of a comprehensive integrated platform for the characterization of a wide range of biotherapeutics, including peptibodies, monoclonal antibodies, and bispecific antibodies. Empowered by this automated IA-CE-MS approach, implementing biotransformation studies at an early drug discovery stage can speed up the drug development process.
Collapse
|
13
|
Arya V, Venkatakrishnan K. Role of Physiologically Based Pharmacokinetic Modeling and Simulation in Enabling Model-Informed Development of Drugs and Biotherapeutics. J Clin Pharmacol 2020; 60 Suppl 1:S7-S11. [PMID: 33205427 DOI: 10.1002/jcph.1770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Vikram Arya
- Division of Infectious Disease Pharmacology (DIDP), Office of Clinical Pharmacology (OCP), Office of Translational Sciences (OTS), Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Karthik Venkatakrishnan
- EMD Serono Research and Development Institute, Inc. (a business of Merck KGaA, Darmstadt, Germany), Billerica, Massachusetts, USA
| |
Collapse
|
14
|
Yu AM, Choi YH, Tu MJ. RNA Drugs and RNA Targets for Small Molecules: Principles, Progress, and Challenges. Pharmacol Rev 2020; 72:862-898. [PMID: 32929000 PMCID: PMC7495341 DOI: 10.1124/pr.120.019554] [Citation(s) in RCA: 229] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
RNA-based therapies, including RNA molecules as drugs and RNA-targeted small molecules, offer unique opportunities to expand the range of therapeutic targets. Various forms of RNAs may be used to selectively act on proteins, transcripts, and genes that cannot be targeted by conventional small molecules or proteins. Although development of RNA drugs faces unparalleled challenges, many strategies have been developed to improve RNA metabolic stability and intracellular delivery. A number of RNA drugs have been approved for medical use, including aptamers (e.g., pegaptanib) that mechanistically act on protein target and small interfering RNAs (e.g., patisiran and givosiran) and antisense oligonucleotides (e.g., inotersen and golodirsen) that directly interfere with RNA targets. Furthermore, guide RNAs are essential components of novel gene editing modalities, and mRNA therapeutics are under development for protein replacement therapy or vaccination, including those against unprecedented severe acute respiratory syndrome coronavirus pandemic. Moreover, functional RNAs or RNA motifs are highly structured to form binding pockets or clefts that are accessible by small molecules. Many natural, semisynthetic, or synthetic antibiotics (e.g., aminoglycosides, tetracyclines, macrolides, oxazolidinones, and phenicols) can directly bind to ribosomal RNAs to achieve the inhibition of bacterial infections. Therefore, there is growing interest in developing RNA-targeted small-molecule drugs amenable to oral administration, and some (e.g., risdiplam and branaplam) have entered clinical trials. Here, we review the pharmacology of novel RNA drugs and RNA-targeted small-molecule medications, with a focus on recent progresses and strategies. Challenges in the development of novel druggable RNA entities and identification of viable RNA targets and selective small-molecule binders are discussed. SIGNIFICANCE STATEMENT: With the understanding of RNA functions and critical roles in diseases, as well as the development of RNA-related technologies, there is growing interest in developing novel RNA-based therapeutics. This comprehensive review presents pharmacology of both RNA drugs and RNA-targeted small-molecule medications, focusing on novel mechanisms of action, the most recent progress, and existing challenges.
Collapse
MESH Headings
- Aptamers, Nucleotide/pharmacology
- Aptamers, Nucleotide/therapeutic use
- Betacoronavirus
- COVID-19
- Chemistry Techniques, Analytical/methods
- Chemistry Techniques, Analytical/standards
- Clustered Regularly Interspaced Short Palindromic Repeats
- Coronavirus Infections/drug therapy
- Drug Delivery Systems/methods
- Drug Development/organization & administration
- Drug Discovery
- Humans
- MicroRNAs/pharmacology
- MicroRNAs/therapeutic use
- Oligonucleotides, Antisense/pharmacology
- Oligonucleotides, Antisense/therapeutic use
- Pandemics
- Pneumonia, Viral/drug therapy
- RNA/adverse effects
- RNA/drug effects
- RNA/pharmacology
- RNA, Antisense/pharmacology
- RNA, Antisense/therapeutic use
- RNA, Messenger/drug effects
- RNA, Messenger/pharmacology
- RNA, Ribosomal/drug effects
- RNA, Ribosomal/pharmacology
- RNA, Small Interfering/pharmacology
- RNA, Small Interfering/therapeutic use
- RNA, Viral/drug effects
- Ribonucleases/metabolism
- Riboswitch/drug effects
- SARS-CoV-2
Collapse
Affiliation(s)
- Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| | - Young Hee Choi
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| | - Mei-Juan Tu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, California (A.-M.Y., Y.H.C., M.-J.T.) and College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyonggi-do, Republic of Korea (Y.H.C.)
| |
Collapse
|