1
|
Roe AL, Krzykwa J, Calderón AI, Bascoul C, Gurley BJ, Koturbash I, Li AP, Liu Y, Mitchell CA, Oketch-Rabah H, Si L, van Breemen RB, Walker H, Ferguson SS. Developing a Screening Strategy to Identify Hepatotoxicity and Drug Interaction Potential of Botanicals. J Diet Suppl 2024; 22:162-192. [PMID: 39450425 DOI: 10.1080/19390211.2024.2417679] [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] [Indexed: 10/26/2024]
Abstract
Botanical supplements, herbal remedies, and plant-derived products are used globally. However, botanical dietary supplements are rarely subjected to robust safety testing unless there are adverse reports in post-market surveillance. Botanicals are complex and difficult to assess using current frameworks designed for single constituent substances (e.g. small molecules or discrete chemicals), making safety assessments costly and time-consuming. The liver is a primary organ of concern for potential botanical-induced hepatotoxicity and botanical-drug interactions as it plays a crucial role in xenobiotic metabolism. The NIH-funded Drug Induced Liver Injury Network noted that the number of botanical-induced liver injuries in 2017 nearly tripled from those observed in 2004-2005. New approach methodologies (NAMs) can aid in the rapid and cost-effective assessment of botanical supplements for potential hepatotoxicity. The Hepatotoxicity Working Group within the Botanical Safety Consortium is working to develop a screening strategy that can help reliably identify potential hepatotoxic botanicals and inform mechanisms of toxicity. This manuscript outlines the Hepatotoxicity Working Group's strategy and describes the assays selected and the rationale for the selection of botanicals used in case studies. The selected NAMs evaluated as a part of this effort are intended to be incorporated into a larger battery of assays to evaluate multiple endpoints related to botanical safety. This work will contribute to a botanical safety toolkit, providing researchers with tools to better understand hepatotoxicity associated with botanicals, prioritize and plan future testing as needed, and gain a deeper insight into the botanicals being tested.
Collapse
Affiliation(s)
- Amy L Roe
- Procter & Gamble Healthcare, Cincinnati, OH, USA
| | - Julie Krzykwa
- Health and Environmental Sciences Institute, Washington, DC, USA
| | - Angela I Calderón
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
| | - Cécile Bascoul
- Product Safety, dōTERRA International, Pleasant Grove, UT, USA
| | - Bill J Gurley
- National Center for Natural Products Research, School of Pharmacy, University of MS, University, MS, USA
| | - Igor Koturbash
- Department of Environmental and Occupational Health, for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | | | - Yitong Liu
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, U.S. Food and Drug Administration, Laurel, MD, USA
| | | | - Hellen Oketch-Rabah
- Office of Dietary Supplement Programs, Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Lin Si
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, USA
- Department of Chemistry, Auburn University at Montgomery, Montgomery, AL, USA
| | - Richard B van Breemen
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR, USA
| | | | - Stephen S Ferguson
- Division of Translational Toxicology, National Institute of Environmental Health Sciences, Durham, NC, USA
| |
Collapse
|
2
|
Liu P, Li Q, Zhu G, Zhang T, Tu D, Zhang F, Finel M, He Y, Ge G. Characterization of the glucuronidating pathway of pectolinarigenin, the major active constituent of the Chinese medicine Daji, in humans and its influence on biological activities. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117280. [PMID: 37797876 DOI: 10.1016/j.jep.2023.117280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/13/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Chinese medicine Daji (the aerial part of Cirsium japonicum DC.) and its charred product (Cirsii Japonici Herba Carbonisata) have been widely used as hemostatic agents or diuretic agents to prepare a variety of Chinese herbal formula. Pectolinarigenin (PEC), one of the most abundant constituents in both Daji and its charred product, has been considered as the key effective substance responsible for the major pharmacological activities of Daji, including hemostasis, hepatoprotective, anti-tumor and anti-osteoporosis effects. However, the major metabolic pathways of PEC in humans and the influence of PEC metabolism on its biological activities are poorly understood. AIM OF THE STUDY To characterize the main metabolic pathway(s) and key enzymes of PEC in human biological systems, as well as to reveal the influence of PEC metabolism on its biological activities. MATERIALS AND METHODS The metabolic stability assays of PEC were investigated in human liver microsomes (HLM). The O-glucuronide of PEC was biosynthesized and characterized by nuclear magnetic resonance (NMR) spectroscopy. The key enzymes responsible for O-glucuronidation of PEC in humans were assigned by performing UGT reaction phenotyping, chemical inhibition and enzymatic kinetic assays. The agonist effects of PEC and its O-glucuronide on nuclear factor erythroid2-related factor 2 (Nrf2), Peroxisome proliferator activated receptors (PPARα and PPARβ) were tested at the cellular level. RESULTS PEC could be readily metabolized to form a mono-O-glucuronide in both human liver microsome (HLM) and human intestinal microsome (HIM). The mono-O-glucuronide was bio-synthesized by mouse liver S9 and its structure was fully characterized as PEC-7-O-β-D-glucuronide (PEC-O-7-G). UGT1A1, UGT1A3 and UGT1A9 are key enzymes responsible for PEC-7-O-glucuronidation in HLM, while UGT1A1, UGT1A9 and 1A10 may play key roles in this reaction in HIM. Biological tests revealed that PEC displayed strong agonist effects on Nrf2, PPARα and PPARβ, whereas PEC-7-O-glucuronide showed relatively weak Nrf2 agonist effect and very weak PPAR agonist effects, indicating that PEC-7-O-glucuronidation strongly weaken its agonist effects on Nrf2 and PPAR. CONCLUSIONS Our results demonstrate that 7-O-glucuronidation is the major metabolic pathway of PEC in human tissues, while UGT1A1, 1A3 and 1A9 are key contributing enzymes responsible for PEC-7-O-glucuronidation in human liver. It is also found that PEC 7-O-glucuronidation significantly weakens the Nrf2 and PPAR agonist effects. All these findings are very helpful for the pharmacologists to deep understand the metabolic rates of PEC in humans.
Collapse
Affiliation(s)
- Peiqi Liu
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qian Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Guanghao Zhu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tiantian Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Dongzhu Tu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Feng Zhang
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Moshe Finel
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, 00014, Finland
| | - Yuqi He
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China.
| | - Guangbo Ge
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
3
|
Li W, Che YN, Chen YY, Wang Z, Wang Z, Jiang LL, Shi HC, Liu Y. Inhibition of UGT1A1*1 and UGT1A1*6 catalyzed glucuronidation of SN-38 by silybins. Chem Biol Interact 2022; 368:110248. [DOI: 10.1016/j.cbi.2022.110248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/23/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
|
4
|
Järvinen E, Deng F, Kiander W, Sinokki A, Kidron H, Sjöstedt N. The Role of Uptake and Efflux Transporters in the Disposition of Glucuronide and Sulfate Conjugates. Front Pharmacol 2022; 12:802539. [PMID: 35095509 PMCID: PMC8793843 DOI: 10.3389/fphar.2021.802539] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/06/2021] [Indexed: 12/11/2022] Open
Abstract
Glucuronidation and sulfation are the most typical phase II metabolic reactions of drugs. The resulting glucuronide and sulfate conjugates are generally considered inactive and safe. They may, however, be the most prominent drug-related material in the circulation and excreta of humans. The glucuronide and sulfate metabolites of drugs typically have limited cell membrane permeability and subsequently, their distribution and excretion from the human body requires transport proteins. Uptake transporters, such as organic anion transporters (OATs and OATPs), mediate the uptake of conjugates into the liver and kidney, while efflux transporters, such as multidrug resistance proteins (MRPs) and breast cancer resistance protein (BCRP), mediate expulsion of conjugates into bile, urine and the intestinal lumen. Understanding the active transport of conjugated drug metabolites is important for predicting the fate of a drug in the body and its safety and efficacy. The aim of this review is to compile the understanding of transporter-mediated disposition of phase II conjugates. We review the literature on hepatic, intestinal and renal uptake transporters participating in the transport of glucuronide and sulfate metabolites of drugs, other xenobiotics and endobiotics. In addition, we provide an update on the involvement of efflux transporters in the disposition of glucuronide and sulfate metabolites. Finally, we discuss the interplay between uptake and efflux transport in the intestine, liver and kidneys as well as the role of transporters in glucuronide and sulfate conjugate toxicity, drug interactions, pharmacogenetics and species differences.
Collapse
Affiliation(s)
- Erkka Järvinen
- Clinical Pharmacology, Pharmacy, and Environmental Medicine, Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Feng Deng
- Department of Clinical Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wilma Kiander
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alli Sinokki
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Heidi Kidron
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Noora Sjöstedt
- Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Cox EJ, Tian DD, Clarke JD, Rettie AE, Unadkat JD, Thummel KE, McCune JS, Paine MF. Modeling Pharmacokinetic Natural Product-Drug Interactions for Decision-Making: A NaPDI Center Recommended Approach. Pharmacol Rev 2021; 73:847-859. [PMID: 33712517 PMCID: PMC7956993 DOI: 10.1124/pharmrev.120.000106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The popularity of botanical and other purported medicinal natural products (NPs) continues to grow, especially among patients with chronic illnesses and patients managed on complex prescription drug regimens. With few exceptions, the risk of a given NP to precipitate a clinically significant pharmacokinetic NP-drug interaction (NPDI) remains understudied or unknown. Application of static or dynamic mathematical models to predict and/or simulate NPDIs can provide critical information about the potential clinical significance of these complex interactions. However, methods used to conduct such predictions or simulations are highly variable. Additionally, published reports using mathematical models to interrogate NPDIs are not always sufficiently detailed to ensure reproducibility. Consequently, guidelines are needed to inform the conduct and reporting of these modeling efforts. This recommended approach from the Center of Excellence for Natural Product Drug Interaction Research describes a systematic method for using mathematical models to interpret the interaction risk of NPs as precipitants of potential clinically significant pharmacokinetic NPDIs. A framework for developing and applying pharmacokinetic NPDI models is presented with the aim of promoting accuracy, reproducibility, and generalizability in the literature. SIGNIFICANCE STATEMENT: Many natural products (NPs) contain phytoconstituents that can increase or decrease systemic or tissue exposure to, and potentially the efficacy of, a pharmaceutical drug; however, no regulatory agency guidelines exist to assist in predicting the risk of these complex interactions. This recommended approach from a multi-institutional consortium designated by National Institutes of Health as the Center of Excellence for Natural Product Drug Interaction Research provides a framework for modeling pharmacokinetic NP-drug interactions.
Collapse
Affiliation(s)
- Emily J Cox
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Dan-Dan Tian
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - John D Clarke
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Allan E Rettie
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Jashvant D Unadkat
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Kenneth E Thummel
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Jeannine S McCune
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Mary F Paine
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (J.D.C., A.E.R., J.D.U., K.E.T., J.S.M., M.F.P.); Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (E.J.C., D.-D.T., J.D.C., M.F.P.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (J.D.U., K.E.T.), University of Washington, Seattle, Washington; and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| |
Collapse
|
6
|
Tanna RS, Tian DD, Cech NB, Oberlies NH, Rettie AE, Thummel KE, Paine MF. Refined Prediction of Pharmacokinetic Kratom-Drug Interactions: Time-Dependent Inhibition Considerations. J Pharmacol Exp Ther 2021; 376:64-73. [PMID: 33093187 PMCID: PMC7745086 DOI: 10.1124/jpet.120.000270] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Preparations from the leaves of the kratom plant (Mitragyna speciosa) are consumed for their opioid-like effects. Several deaths have been associated with kratom used concomitantly with some drugs. Pharmacokinetic interactions are potential underlying mechanisms of these fatalities. Accumulating in vitro evidence has demonstrated select kratom alkaloids, including the abundant indole alkaloid mitragynine, as reversible inhibitors of several cytochromes P450 (CYPs). The objective of this work was to refine the mechanistic understanding of potential kratom-drug interactions by considering both reversible and time-dependent inhibition (TDI) of CYPs in the liver and intestine. Mitragynine was tested against CYP2C9 (diclofenac 4'-hydroxylation), CYP2D6 (dextromethorphan O-demethylation), and CYP3A (midazolam 1'-hydroxylation) activities in human liver microsomes (HLMs) and CYP3A activity in human intestinal microsomes (HIMs). Comparing the absence to presence of NADPH during preincubation of mitragynine with HLMs or HIMs, an ∼7-fold leftward shift in IC50 (∼20 to 3 μM) toward CYP3A resulted, prompting determination of TDI parameters (HLMs: K I , 4.1 ± 0.9 μM; k inact , 0.068 ± 0.01 min-1; HIMs: K I , 4.2 ± 2.5 μM; k inact , 0.079 ± 0.02 min-1). Mitragynine caused no leftward shift in IC50 toward CYP2C9 (∼40 μM) and CYP2D6 (∼1 μM) but was a strong competitive inhibitor of CYP2D6 (K i , 1.17 ± 0.07 μM). Using a recommended mechanistic static model, mitragynine (2-g kratom dose) was predicted to increase dextromethorphan and midazolam area under the plasma concentration-time curve by 1.06- and 5.69-fold, respectively. The predicted midazolam area under the plasma concentration-time curve ratio exceeded the recommended cutoff (1.25), which would have been missed if TDI was not considered. SIGNIFICANCE STATEMENT: Kratom, a botanical natural product increasingly consumed for its opioid-like effects, may precipitate potentially serious pharmacokinetic interactions with drugs. The abundant kratom indole alkaloid mitragynine was shown to be a time-dependent inhibitor of hepatic and intestinal cytochrome P450 3A activity. A mechanistic static model predicted mitragynine to increase systemic exposure to the probe drug substrate midazolam by 5.7-fold, necessitating further evaluation via dynamic models and clinical assessment to advance the understanding of consumer safety associated with kratom use.
Collapse
Affiliation(s)
- Rakshit S Tanna
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Dan-Dan Tian
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Nadja B Cech
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Nicholas H Oberlies
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Allan E Rettie
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Kenneth E Thummel
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| | - Mary F Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (R.S.T., D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (N.B.C., N.H.O.); Departments of Medicinal Chemistry (A.E.R.) and Pharmaceutics (K.E.T.), School of Pharmacy, University of Washington, Seattle, Washington; and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., A.E.R., K.E.T., M.F.P.)
| |
Collapse
|
7
|
Pharmacokinetic Interactions between Herbal Medicines and Drugs: Their Mechanisms and Clinical Relevance. Life (Basel) 2020; 10:life10070106. [PMID: 32635538 PMCID: PMC7400069 DOI: 10.3390/life10070106] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 01/20/2023] Open
Abstract
The therapeutic efficacy of a drug or its unexpected unwanted side effects may depend on the concurrent use of a medicinal plant. In particular, constituents in the medicinal plant extracts may influence drug bioavailability, metabolism and half-life, leading to drug toxicity or failure to obtain a therapeutic response. This narrative review focuses on clinical studies improving knowledge on the ability of selected herbal medicines to influence the pharmacokinetics of co-administered drugs. Moreover, in vitro studies are useful to anticipate potential herbal medicine-drug interactions. In particular, they help to elucidate the cellular target (metabolic or transporter protein) and the mechanism (induction or inhibition) by which a single constituent of the herbal medicine acts. The authors highlight the difficulties in predicting herbal–drug interactions from in vitro data where high concentrations of extracts or their constituents are used and pharmacokinetics are missed. Moreover, the difficulty to compare results from human studies where different kinds of herbal extracts are used is discussed. The herbal medicines discussed are among the best sellers and they are reported in the “Herbal Medicines for Human Use” section of the European Medicinal Agency (EMA).
Collapse
|
8
|
Kellogg JJ, Paine MF, McCune JS, Oberlies NH, Cech NB. Selection and characterization of botanical natural products for research studies: a NaPDI center recommended approach. Nat Prod Rep 2019; 36:1196-1221. [PMID: 30681109 PMCID: PMC6658353 DOI: 10.1039/c8np00065d] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Covering: up to the end of 2018 Dietary supplements, which include botanical (plant-based) natural products, constitute a multi-billion-dollar industry in the US. Regulation and quality control for this industry is an ongoing challenge. While there is general agreement that rigorous scientific studies are needed to evaluate the safety and efficacy of botanical natural products used by consumers, researchers conducting such studies face a unique set of challenges. Botanical natural products are inherently complex mixtures, with composition that differs depending on myriad factors including variability in genetics, cultivation conditions, and processing methods. Unfortunately, many studies of botanical natural products are carried out with poorly characterized study material, such that the results are irreproducible and difficult to interpret. This review provides recommended approaches for addressing the critical questions that researchers must address prior to in vitro or in vivo (including clinical) evaluation of botanical natural products. We describe selection and authentication of botanical material and identification of key biologically active compounds, and compare state-of-the-art methodologies such as untargeted metabolomics with more traditional targeted methods of characterization. The topics are chosen to be of maximal relevance to researchers, and are reviewed critically with commentary as to which approaches are most practical and useful and what common pitfalls should be avoided.
Collapse
Affiliation(s)
- Joshua J. Kellogg
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA.
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Jeannine S. McCune
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Nicholas H. Oberlies
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA.
| | - Nadja B. Cech
- Department of Chemistry & Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA.
| |
Collapse
|
9
|
Multiple circulating saponins from intravenous ShenMai inhibit OATP1Bs in vitro: potential joint precipitants of drug interactions. Acta Pharmacol Sin 2019; 40:833-849. [PMID: 30327544 DOI: 10.1038/s41401-018-0173-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 09/14/2018] [Indexed: 11/08/2022]
Abstract
ShenMai, an intravenous injection prepared from steamed Panax ginseng roots (Hongshen) and Ophiopogon japonicus roots (Maidong), is used as an add-on therapy for coronary artery disease and cancer; saponins are its bioactive constituents. Since many saponins inhibit human organic anion-transporting polypeptides (OATP)1B, this investigation determined the inhibition potencies of circulating ShenMai saponins on the transporters and the joint potential of these compounds for ShenMai-drug interaction. Circulating saponins and their pharmacokinetics were characterized in rats receiving a 30-min infusion of ShenMai at 10 mL/kg. Inhibition of human OATP1B1/1B3 and rat Oatp1b2 by the individual saponins was investigated in vitro; the compounds' joint inhibition was also assessed in vitro and the data was processed using the Chou-Talalay method. Plasma protein binding was assessed by equilibrium dialysis. Altogether, 49 saponins in ShenMai were characterized and graded into: 10-100 μmol/day (compound doses from ShenMai; 7 compounds), 1-10 μmol/day (17 compounds), and <1 μmol/day (25 compounds, including Maidong ophiopogonins). After dosing, circulating saponins were protopanaxadiol-type ginsenosides Rb1, Rb2, Rc, Rd, Ra1, Rg3, Ra2, and Ra3, protopanaxatriol-type ginsenosides Rg1, Re, Rg2, and Rf, and ginsenoside Ro. The protopanaxadiol-type ginsenosides exhibited maximum plasma concentrations of 2.1-46.6 μmol/L, plasma unbound fractions of 0.4-1.0% and terminal half-lives of 15.6-28.5 h (ginsenoside Rg3, 1.9 h), while the other ginsenosides exhibited 0.1-7.7 μmol/L, 20.8-99.2%, and 0.2-0.5 h, respectively. The protopanaxadiol-type ginsenosides, ginsenosides without any sugar attachment at C-20 (except ginsenoside Rf), and ginsenoside Ro inhibited OATP1B3 more potently (IC50, 0.2-3.5 µmol/L) than the other ginsenosides (≥22.6 µmol/L). Inhibition of OATP1B1 by ginsenosides was less potent than OATP1B3 inhibition. Ginsenosides Rb1, Rb2, Rc, Rd, Ro, Ra1, Re, and Rg2 likely contribute the major part of OATP1B3-mediated ShenMai-drug interaction potential, in an additive and time-related manner.
Collapse
|
10
|
Li Y, Wu Y, Li YJ, Meng L, Ding CY, Dong ZJ. Effects of Silymarin on the In Vivo Pharmacokinetics of Simvastatin and Its Active Metabolite in Rats. MOLECULES (BASEL, SWITZERLAND) 2019; 24:molecules24091666. [PMID: 31035343 PMCID: PMC6540003 DOI: 10.3390/molecules24091666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/23/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
Herein, the effect of silymarin pretreatment on the pharmacokinetics of simvastatin in rats was evaluated. To ensure the accuracy of the results, a rapid and sensitive UPLC-MS/MS method was established for simultaneous quantification of simvastatin (SV) and its active metabolite simvastatin acid (SVA). This method was applied for studying the pharmacokinetic interactions in rats after oral co-administration of silymarin (45 mg/kg) and different concentrations of SV. The major pharmacokinetic parameters, including Cmax, tmax, t1/2, mean residence time (MRT), elimination rate constant (λz) and area under the concentration-time curve (AUC0-12h), were calculated using the non-compartmental model. The results showed that the co-administration of silymarin and SV significantly increased the Cmax and AUC0-12h of SVA compared with SV alone, while there was no significant difference with regards to Tmax and t1/2. However, SV pharmacokinetic parameters were not significantly affected by silymarin pretreatment. Therefore, these changes indicated that drug-drug interactions may occur after co-administration of silymarin and SV.
Collapse
Affiliation(s)
- Ying Li
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| | - Yin Wu
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| | - Ya-Jing Li
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| | - Lu Meng
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| | - Cong-Yang Ding
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| | - Zhan-Jun Dong
- National Clinical Drug Monitoring Center, Department of Pharmacy, Hebei Province General Center, Shijiazhuang 050051, China.
| |
Collapse
|
11
|
The mechanisms of pharmacokinetic food-drug interactions - A perspective from the UNGAP group. Eur J Pharm Sci 2019; 134:31-59. [PMID: 30974173 DOI: 10.1016/j.ejps.2019.04.003] [Citation(s) in RCA: 206] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/12/2019] [Accepted: 04/02/2019] [Indexed: 02/06/2023]
Abstract
The simultaneous intake of food and drugs can have a strong impact on drug release, absorption, distribution, metabolism and/or elimination and consequently, on the efficacy and safety of pharmacotherapy. As such, food-drug interactions are one of the main challenges in oral drug administration. Whereas pharmacokinetic (PK) food-drug interactions can have a variety of causes, pharmacodynamic (PD) food-drug interactions occur due to specific pharmacological interactions between a drug and particular drinks or food. In recent years, extensive efforts were made to elucidate the mechanisms that drive pharmacokinetic food-drug interactions. Their occurrence depends mainly on the properties of the drug substance, the formulation and a multitude of physiological factors. Every intake of food or drink changes the physiological conditions in the human gastrointestinal tract. Therefore, a precise understanding of how different foods and drinks affect the processes of drug absorption, distribution, metabolism and/or elimination as well as formulation performance is important in order to be able to predict and avoid such interactions. Furthermore, it must be considered that beverages such as milk, grapefruit juice and alcohol can also lead to specific food-drug interactions. In this regard, the growing use of food supplements and functional food requires urgent attention in oral pharmacotherapy. Recently, a new consortium in Understanding Gastrointestinal Absorption-related Processes (UNGAP) was established through COST, a funding organisation of the European Union supporting translational research across Europe. In this review of the UNGAP Working group "Food-Drug Interface", the different mechanisms that can lead to pharmacokinetic food-drug interactions are discussed and summarised from different expert perspectives.
Collapse
|
12
|
Waidyanatha S, Ryan K, Roe AL, Jia W, Paine MF, Ferguson S, Gurley BJ, Welch K, Chow MSS, Devito M, Rider C. Follow that botanical: Challenges and recommendations for assessing absorption, distribution, metabolism and excretion of botanical dietary supplements. Food Chem Toxicol 2018; 121:194-202. [PMID: 30170118 DOI: 10.1016/j.fct.2018.08.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 08/10/2018] [Accepted: 08/24/2018] [Indexed: 12/01/2022]
Abstract
Botanical dietary supplements are complex mixtures containing one or more botanical ingredient(s), each containing numerous constituents potentially responsible for its purported biological activity. Absorption, distribution, metabolism, and excretion (ADME) data are critical to understand the safety of botanical dietary supplements, including their potential for pharmacokinetic botanical-drug or botanical-botanical interactions. However, ADME data for botanical dietary supplements are rarely available and frequently inadequate to characterize their fate in vivo. Based on an assessment of the current status of botanical dietary supplements ADME research, the following key areas are identified that require robust data for human safety assessment: 1) phytochemical characterization including contaminant analysis and botanical authentication; 2) in vitro and/or in vivo data for identifying potential botanical-botanical or botanical-drug interactions and active/marker constituents; 3) robust ADME study design to include systemic exposure data on active/marker constituents using traditional or novel analytical chemistry and statistical approaches such as poly-pharmacokinetics; and 4) investigation of human relevance. A case study with Ginkgo biloba extract is used to highlight the challenges and proposed approaches in using ADME data for human safety assessment of botanical dietary supplements.
Collapse
Affiliation(s)
- Suramya Waidyanatha
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA.
| | - Kristen Ryan
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Amy L Roe
- The Procter & Gamble Company, Cincinnati, OH, USA
| | - Wei Jia
- University of Hawaii, Manoa, HI, USA
| | - Mary F Paine
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, WA, USA
| | - Stephen Ferguson
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Bill J Gurley
- University of Arkansas for Medical Sciences, College of Pharmacy, Little Rock, AR, USA
| | - Kevin Welch
- United States Department of Agriculture, Logan, UT, USA
| | - Moses S S Chow
- Western University of Health Sciences, College of Pharmacy, Pomona, CA, USA
| | - Michael Devito
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Cynthia Rider
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| |
Collapse
|
13
|
Tian DD, Kellogg JJ, Okut N, Oberlies NH, Cech NB, Shen DD, McCune JS, Paine MF. Identification of Intestinal UDP-Glucuronosyltransferase Inhibitors in Green Tea ( Camellia sinensis) Using a Biochemometric Approach: Application to Raloxifene as a Test Drug via In Vitro to In Vivo Extrapolation. Drug Metab Dispos 2018; 46:552-560. [PMID: 29467215 PMCID: PMC5890833 DOI: 10.1124/dmd.117.079491] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/14/2018] [Indexed: 11/22/2022] Open
Abstract
Green tea (Camellia sinensis) is a popular beverage worldwide, raising concern for adverse interactions when co-consumed with conventional drugs. Like many botanical natural products, green tea contains numerous polyphenolic constituents that undergo extensive glucuronidation. As such, the UDP-glucuronosyltransferases (UGTs), particularly intestinal UGTs, represent potential first-pass targets for green tea-drug interactions. Candidate intestinal UGT inhibitors were identified using a biochemometrics approach, which combines bioassay and chemometric data. Extracts and fractions prepared from four widely consumed teas were screened (20-180 μg/ml) as inhibitors of UGT activity (4-methylumbelliferone glucuronidation) in human intestinal microsomes; all demonstrated concentration-dependent inhibition. A biochemometrics-identified fraction rich in UGT inhibitors from a representative tea was purified further and subjected to second-stage biochemometric analysis. Five catechins were identified as major constituents in the bioactive subfractions and prioritized for further evaluation. Of these catechins, (-)-epicatechin gallate and (-)-epigallocatechin gallate showed concentration-dependent inhibition, with IC50 values (105 and 59 μM, respectively) near or below concentrations measured in a cup (240 ml) of tea (66 and 240 μM, respectively). Using the clinical intestinal UGT substrate raloxifene, the Ki values were ∼1.0 and 2.0 μM, respectively. Using estimated intestinal lumen and enterocyte inhibitor concentrations, a mechanistic static model predicted green tea to increase the raloxifene plasma area under the curve up to 6.1- and 1.3-fold, respectively. Application of this novel approach, which combines biochemometrics with in vitro-in vivo extrapolation, to other natural product-drug combinations will refine these procedures, informing the need for further evaluation via dynamic modeling and clinical testing.
Collapse
Affiliation(s)
- Dan-Dan Tian
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Joshua J Kellogg
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Neşe Okut
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Nicholas H Oberlies
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Nadja B Cech
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Danny D Shen
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Jeannine S McCune
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| | - Mary F Paine
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington (D.-D.T., M.F.P.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina (J.J.K., N.O., N.H.O., N.B.C.); Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington (D.D.S.); and Department of Population Sciences, City of Hope, Duarte, California (J.S.M.)
| |
Collapse
|
14
|
Grimstein M, Huang SM. A regulatory science viewpoint on botanical-drug interactions. J Food Drug Anal 2018; 26:S12-S25. [PMID: 29703380 PMCID: PMC9326881 DOI: 10.1016/j.jfda.2018.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/20/2018] [Accepted: 01/23/2018] [Indexed: 11/28/2022] Open
Abstract
There is a continued predisposition of concurrent use of drugs and botanical products. Consumers often self-administer botanical products without informing their health care providers. The perceived safety of botanical products with lack of knowledge of the interaction potential poses a challenge for providers and both efficacy and safety concerns for patients. Botanical–drug combinations can produce untoward effects when botanical constituents modulate drug metabolizing enzymes and/or transporters impacting the systemic or tissue exposure of concomitant drugs. Examples of pertinent scientific literature evaluating the interaction potential of commonly used botanicals in the US are discussed. Current methodologies that can be applied to advance our efforts in predicting drug interaction liability is presented. This review also highlights the regulatory science viewpoint on botanical–drug interactions and labeling implications.
Collapse
Affiliation(s)
- Manuela Grimstein
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA.
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
15
|
Necyk C, Zubach-Cassano L. Natural Health Products and Diabetes: A Practical Review. Can J Diabetes 2017; 41:642-647. [PMID: 28826695 DOI: 10.1016/j.jcjd.2017.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 06/27/2017] [Accepted: 06/27/2017] [Indexed: 11/30/2022]
Abstract
The prevalence of natural health product (NHP) use in Canada is extensive. Patients with chronic diseases, including diabetes, use NHPs at a higher rate than the general population. Many NHPs exert hypoglycemic effects, among other effects relevant to diabetes management. To provide a practical, clinical review of NHPs with such effects targeted to pharmacists, a literature search was performed to collect data on the efficacy and safety profiles of 10 commonly used NHPs that exert antidiabetic properties. The following NHPs are included in this clinical review: alpha-lipoic acid, chromium, magnesium, bitter melon, cinnamon, fenugreek, gymnema, milk thistle, Reishi mushroom and white mulberry. Given the potential of NHPs to additively cause hypoglycemia when used concurrently with conventional medications, pharmacists should be up to date with current evidence around NHPs that may affect diabetes care to prevent adverse reactions and interactions. In addition, effective and respectful communication with patients around NHP use and collaboration with various health-care providers are essential in the patient care process.
Collapse
Affiliation(s)
- Candace Necyk
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| | | |
Collapse
|
16
|
|
17
|
Roe AL, Paine MF, Gurley BJ, Brouwer KR, Jordan S, Griffiths JC. Assessing Natural Product-Drug Interactions: An End-to-End Safety Framework. Regul Toxicol Pharmacol 2016; 76:1-6. [PMID: 26776752 DOI: 10.1016/j.yrtph.2016.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/28/2022]
Abstract
The use of natural products (NPs), including herbal medicines and other dietary supplements, by North Americans continues to increase across all age groups. This population has access to conventional medications, with significant polypharmacy observed in older adults. Thus, the safety of the interactions between multi-ingredient NPs and drugs is a topic of paramount importance. Considerations such as history of safe use, literature data from animal toxicity and human clinical studies, and NP constituent characterization would provide guidance on whether to assess NP-drug interactions experimentally. The literature is replete with reports of various NP extracts and constituents as potent inhibitors of drug metabolizing enzymes, and transporters. However, without standard methods for NP characterization or in vitro testing, extrapolating these reports to clinically-relevant NP-drug interactions is difficult. This lack of a clear definition of risk precludes clinicians and consumers from making informed decisions about the safety of taking NPs with conventional medications. A framework is needed that describes an integrated robust approach for assessing NP-drug interactions; and, translation of the data into formulation alterations, dose adjustment, labelling, and/or post-marketing surveillance strategies. A session was held at the 41st Annual Summer Meeting of the Toxicology Forum in Colorado Springs, CO, to highlight the challenges and critical components that should be included in a framework approach.
Collapse
Affiliation(s)
- Amy L Roe
- Product Safety & Regulatory Affairs, The Procter & Gamble Company, Cincinnati, OH 45040, United States.
| | - Mary F Paine
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, WA 99210, United States.
| | - Bill J Gurley
- College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States.
| | | | - Scott Jordan
- Marketed Biologicals, Biotechnology and Natural Health Products Bureau, Marketed Health Products Directorate, Health Canada, Ottawa, Ontario, Canada.
| | - James C Griffiths
- Council for Responsible Nutrition, Washington, DC 20036, United States.
| |
Collapse
|
18
|
Gufford BT, Barr JT, González-Pérez V, Layton ME, White JR, Oberlies NH, Paine MF. Quantitative prediction and clinical evaluation of an unexplored herb-drug interaction mechanism in healthy volunteers. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:701-10. [PMID: 26904384 PMCID: PMC4759704 DOI: 10.1002/psp4.12047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Quantitative prediction of herb–drug interaction risk remains challenging. A quantitative framework to assess a potential interaction was used to evaluate a mechanism not previously tested in humans. The semipurified milk thistle product, silibinin, was selected as an exemplar herbal product inhibitor of raloxifene intestinal glucuronidation. Physiologically based pharmacokinetic (PBPK) model simulations of the silibinin–raloxifene interaction predicted up to 30% increases in raloxifene area under the curve (AUC0‐inf) and maximal concentration (Cmax). Model‐informed clinical evaluation of the silibinin–raloxifene interaction indicated minimal clinical interaction liability, with observed geometric mean raloxifene AUC0‐inf and Cmax ratios lying within the predefined no effect range (0.75–1.33). Further refinement of PBPK modeling and simulation approaches will enhance confidence in predictions and facilitate generalizability to additional herb–drug combinations. This quantitative framework can be used to develop guidances to evaluate potential herb–drug interactions prospectively, providing evidenced‐based information about the risk or safety of these interactions.
Collapse
Affiliation(s)
- B T Gufford
- College of Pharmacy Washington State University Spokane, Washington USA
| | - J T Barr
- College of Pharmacy Washington State University Spokane, Washington USA
| | - V González-Pérez
- College of Pharmacy Washington State University Spokane, Washington USA
| | - M E Layton
- College of Medical Sciences Washington State University Spokane, Washington USA
| | - J R White
- College of Pharmacy Washington State University Spokane, Washington USA
| | - N H Oberlies
- Department of Chemistry and Biochemistry University of North Carolina at Greensboro Greensboro North Carolina USA
| | - M F Paine
- College of Pharmacy Washington State University Spokane, Washington USA
| |
Collapse
|
19
|
Gufford BT, Graf TN, Paguigan ND, Oberlies NH, Paine MF. Chemoenzymatic Synthesis, Characterization, and Scale-Up of Milk Thistle Flavonolignan Glucuronides. Drug Metab Dispos 2015; 43:1734-43. [PMID: 26316643 PMCID: PMC4613946 DOI: 10.1124/dmd.115.066076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/26/2015] [Indexed: 11/22/2022] Open
Abstract
Plant-based therapeutics, including herbal products, continue to represent a growing facet of the contemporary health care market. Mechanistic descriptions of the pharmacokinetics and pharmacodynamics of constituents composing these products remain nascent, particularly for metabolites produced following herbal product ingestion. Generation and characterization of authentic metabolite standards are essential to improve the quantitative mechanistic understanding of herbal product disposition in both in vitro and in vivo systems. Using the model herbal product, milk thistle, the objective of this work was to biosynthesize multimilligram quantities of glucuronides of select constituents (flavonolignans) to fill multiple knowledge gaps in the understanding of herbal product disposition and action. A partnership between clinical pharmacology and natural products chemistry expertise was leveraged to optimize reaction conditions for efficient glucuronide formation and evaluate alternate enzyme and reagent sources to improve cost effectiveness. Optimized reaction conditions used at least one-fourth the amount of microsomal protein (from bovine liver) and cofactor (UDP glucuronic acid) compared with typical conditions using human-derived subcellular fractions, providing substantial cost savings. Glucuronidation was flavonolignan-dependent. Silybin A, silybin B, isosilybin A, and isosilybin B generated five, four, four, and three monoglucuronides, respectively. Large-scale synthesis (40 mg of starting material) generated three glucuronides of silybin A: silybin A-7-O-β-D-glucuronide (15.7 mg), silybin A-5-O-β-D-glucuronide (1.6 mg), and silybin A-4´´-O-β-D-glucuronide (11.1 mg). This optimized, cost-efficient method lays the foundation for a systematic approach to synthesize and characterize herbal product constituent glucuronides, enabling an improved understanding of mechanisms underlying herbal product disposition and action.
Collapse
Affiliation(s)
- Brandon T Gufford
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (B.T.G., M.F.P.); and Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, North Carolina (T.N.G., N.D.P., N.H.O.)
| | - Tyler N Graf
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (B.T.G., M.F.P.); and Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, North Carolina (T.N.G., N.D.P., N.H.O.)
| | - Noemi D Paguigan
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (B.T.G., M.F.P.); and Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, North Carolina (T.N.G., N.D.P., N.H.O.)
| | - Nicholas H Oberlies
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (B.T.G., M.F.P.); and Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, North Carolina (T.N.G., N.D.P., N.H.O.)
| | - Mary F Paine
- Experimental and Systems Pharmacology, College of Pharmacy, Washington State University, Spokane, Washington (B.T.G., M.F.P.); and Department of Chemistry and Biochemistry, The University of North Carolina at Greensboro, Greensboro, North Carolina (T.N.G., N.D.P., N.H.O.)
| |
Collapse
|