1
|
Nguyen JT, Arian CM, Tanna RS, Cherel MG, Layton ME, White JR, Thummel KE, Paine MF. The Pharmacokinetic Interaction Between Metformin and the Natural Product Goldenseal Is Metformin Dose-Dependent: A Three-Arm Crossover Study in Adults With Type 2 Diabetes. Clin Transl Sci 2025; 18:e70120. [PMID: 39943692 PMCID: PMC11821731 DOI: 10.1111/cts.70120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/02/2024] [Accepted: 12/14/2024] [Indexed: 02/16/2025] Open
Abstract
Pharmacokinetic drug interactions can lead to unexpected changes in plasma concentrations of the object drug, potentially increasing the risk for adverse effects and/or decreasing therapeutic efficacy. The botanical product goldenseal was previously shown to decrease metformin systemic exposure in healthy adults. This three-arm, open-label, crossover clinical study assessed the pharmacokinetic goldenseal-metformin interaction in adults with type 2 diabetes stabilized on therapeutic doses of metformin (500-2550 mg daily). The aggregate pharmacokinetic data indicated no clinically meaningful interaction as determined by the metformin area under the plasma concentration-time curve (AUC) geometric mean ratio [90% confidence interval] of 0.93 [0.86-1.01] laying within the predefined no-effect range (0.80-1.25). However, metformin AUC decreased by ~20%, 14%, and 0% after goldenseal coadministration at low (500-750 mg), moderate (1000-1500 mg), and high (2000-2550 mg) metformin doses, respectively; renal clearance and half-life remained unchanged throughout. The exploratory pharmacodynamic endpoint, HbA1c, decreased on average from 6.8% to 6.5%, regardless of the effects of goldenseal on metformin pharmacokinetics. The decreasing effect of goldenseal on metformin systemic exposure with increasing metformin dose, coupled with no changes in renal excretion and elimination half-life, indicated that both the pharmacokinetic goldenseal-metformin interaction and the nonlinear absorption of metformin are governed by saturable, intestinal transport mechanism(s). The disconnect between changes in metformin systemic exposure and therapeutic effects emphasizes the need to evaluate clinical biomarkers to comprehensively assess drug interaction risks, particularly those involving natural products. Healthcare providers may consider cautioning patients about supplementing metformin pharmacotherapy with goldenseal to avoid risks for undesired changes in glycemic control. Trial Registration: ClinicalTrials.gov identifier: NCT05081583.
Collapse
Affiliation(s)
- James T. Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Christopher M. Arian
- Department of Pharmaceutics, School of PharmacyUniversity of WashingtonSeattleWashingtonUSA
| | - Rakshit S. Tanna
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Maxey G. Cherel
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Matthew E. Layton
- Elson S. Floyd College of MedicineWashington State UniversitySpokaneWashingtonUSA
| | - John R. White
- Department of Pharmacotherapy, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Kenneth E. Thummel
- Department of Pharmaceutics, School of PharmacyUniversity of WashingtonSeattleWashingtonUSA
- Center of Excellence for Natural Product Drug Interaction ResearchSpokaneWashingtonUSA
| | - Mary F. Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
- Center of Excellence for Natural Product Drug Interaction ResearchSpokaneWashingtonUSA
| |
Collapse
|
2
|
Akyüz GY. Impact of antioxidant excipients on N-nitrosamine formation and bioequivalence in metformin formulations (review article). ANNALES PHARMACEUTIQUES FRANÇAISES 2025:S0003-4509(25)00012-4. [PMID: 39892475 DOI: 10.1016/j.pharma.2025.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
The discovery of N-nitrosamine impurities in pharmaceutical products has raised serious quality concerns, particularly in metformin products, which are widely used in the treatment of type 2 diabetes mellitus. The detection of N-nitrosodimethylamine (NDMA) in metformin products has led to global recalls and increased regulatory investigations. Generic manufacturers face the challenge of balancing stringent bioequivalence requirements for Biopharmaceutical Classification System (BCS) Class III drugs, which require strict control of excipient composition while ensuring N-nitrosamine control and therapeutic equivalence. The use of antioxidants as a strategy to reduce N-nitrosamine formation requires careful consideration to maintain both bioequivalence and product safety. This article evaluates the use of antioxidants for the prevention of N-nitrosamine formation in metformin formulations, addressing the implications of this strategy on bioequivalence and its relationship with the regulatory framework.
Collapse
|
3
|
Asaji S, Funai Y, Seki Y, Tamai I, Shirasaka Y. Contributions of multiple transport mechanisms to intestinal uptake of serotonin. J Pharm Sci 2024; 113:3216-3226. [PMID: 39278593 DOI: 10.1016/j.xphs.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 09/18/2024]
Abstract
This study aimed to analyze the contributions of multiple transport mechanisms to the intestinal uptake of serotonin (5-HT) by employing a variety of in vitro experimental techniques, focusing on organic cation transporters expressed in the gastrointestinal (GI) tract, such as SERT, PMAT, THTR2, OCT3, and OCTN2. Analysis of the concentration dependence of 5-HT uptake by Caco-2 cells revealed multi-affinity kinetics with high-affinity and low-affinity components, suggesting that multiple transporters are involved in the intestinal 5-HT uptake. Comparative analysis of transporters using Km values obtained in Xenopus oocyte expression systems suggested that SERT is responsible for the high-affinity transport, while PMAT, THTR2, and OCT3 contribute to the low-affinity transport. Further analysis indicated that the relative contributions of SERT and PMAT to the intestinal 5-HT uptake (0.01 µM) are approximately 94.9% and 1.1%, respectively. Interestingly, at the concentration of 10 µM, the reported steady-state concentration of 5-HT in the human colon, the contributions of SERT, PMAT, THTR2, and OCT3 were estimated to be approximately 37.0%, 1.0%, 18.2%, and 20.5%, respectively. In conclusion, the present study indicated that the contributions of multiple transporters to 5-HT uptake in the GI tract are dependent upon the colon luminal concentration of 5-HT.
Collapse
Affiliation(s)
- Suguru Asaji
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuta Funai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yuta Seki
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Ikumi Tamai
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmacy, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan.
| |
Collapse
|
4
|
Natrus L, Lisakovska O, Smirnov A, Osadchuk Y, Klys Y. Apoptotic and proliferative processes in the small intestine of rats with type 2 diabetes mellitus after metformin and propionic acid treatment. Front Pharmacol 2024; 15:1477793. [PMID: 39478962 PMCID: PMC11521949 DOI: 10.3389/fphar.2024.1477793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Background Propionic acid (PA) is an intermediate product of metabolism of intestinal bacteria and may protect the intestinal barrier from disruption. The aim of the study was to investigate the apoptotic and proliferative processes in the small intestine (SI) of rats with type 2 diabetes mellitus (T2DM) on the background of metformin monotherapy and its combination with PA. Methods Male Wistar rats were divided: 1) control; 2) T2DM (3-month high-fat diet followed by streptozotocin injection of 25 mg/kg of body weight); 3) T2DM + metformin (60 mg/kg, 14 days, orally); 4) T2DM + PA (60 mg/kg, 14 days, orally); 5) T2DM + PA + metformin. Western blotting, RT-PCR, and scanning electron microscopy were performed. Results We observed profound changes in the SI of diabetic rats suggesting the disturbed intestinal homeostasis: impaired mitochondrial ultrastructure, increased cristae volume, and decreased content of proliferative marker Ki67 with almost unchanged proapoptotic caspase-3 and its p17 subunit levels. Metformin and PA monotherapies also led to an increased cristae volume, however, after their combination, a tendency to normalization of ultrastructure of mitochondria was observed. While there was a significant inhibition of proliferation in T2DM and, in greater extent, after metformin and PA monotherapies, differential influence on apoptosis in the SI was observed. While metformin inhibited apoptosis via Bax declining, PA mainly acted via caspase-3-dependent mechanism elevating its active p17 subunit. Conclusion PA supplementation for the improvement of diabetes-induced gastrointestinal complications concurrently with metformin may be consider as a perspective supportive therapy. Data related to PA action on SI may be valuable during the development of new treatment strategies for diabetes-induced intestinal disturbances raised after metformin treatment.
Collapse
Affiliation(s)
- Larysa Natrus
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomolets National Medical University, Kyiv, Ukraine
| | - Olha Lisakovska
- Department of Biochemistry of Vitamins and Coenzymes, Palladin Institute of Biochemistry, Kyiv, Ukraine
| | - Anton Smirnov
- Department of Socio-Humanitarian and Biomedical Sciences, Kharkiv Institute of Medicine and Biomedical Sciences, Kharkiv, Ukraine
| | - Yuliia Osadchuk
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomolets National Medical University, Kyiv, Ukraine
| | - Yuliia Klys
- Department of Modern Technologies of Medical Diagnostics and Treatment, Bogomolets National Medical University, Kyiv, Ukraine
| |
Collapse
|
5
|
Vachalova V, Kumnova F, Synova T, Anandam KY, Abad C, Karahoda R, Staud F. Metformin inhibits OCT3-mediated serotonin transport in the placenta. Biomed Pharmacother 2024; 179:117399. [PMID: 39243433 DOI: 10.1016/j.biopha.2024.117399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
Proper fetal development requires tight regulation of serotonin concentrations within the fetoplacental unit. This homeostasis is partly maintained by the placental transporter OCT3/SLC22A3, which takes up serotonin from the fetal circulation. Metformin, an antidiabetic drug commonly used to treat gestational diabetes mellitus, was shown to inhibit OCT3. We, therefore, hypothesized that its use during pregnancy could disrupt placental serotonin homeostasis. This hypothesis was tested using three experimental model systems: primary trophoblast cells isolated from the human term placenta, fresh villous human term placenta fragments, and rat term placenta perfusions. Inhibition of serotonin transport by metformin at three concentrations (1 μM, 10 μM, and 100 μM) was assessed in all three models. The OCT3 inhibitor decynium-22 (100 μM) and paroxetine (100 μM), a dual inhibitor of SERT and OCT3, were used as controls. In primary trophoblasts, paroxetine exhibited the strongest inhibition of serotonin uptake, followed by decynium-22. Metformin showed a concentration-dependent effect, reducing serotonin uptake by up to 57 % at the highest concentration. Its inhibitory effect was less pronounced in fresh villous fragments but remained statistically significant at all concentrations. In the perfused rat placenta, metformin demonstrated a concentration-dependent effect, reducing placental serotonin uptake by 44 % at the highest concentration tested. Our findings across all experimental models show inhibition of placental OCT3 by metformin, resulting in reduced serotonin uptake by the trophoblast. This sheds light on mechanisms that may underpin metformin-mediated effects on fetal development.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Fiona Kumnova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
6
|
Camarero-Hoyos C, Bouzón-Arnáiz I, Avalos-Padilla Y, Fallica AN, Román-Álamo L, Ramírez M, Portabella E, Cuspinera O, Currea-Ayala D, Orozco-Quer M, Ribera M, Siden-Kiamos I, Spanos L, Iglesias V, Crespo B, Viera S, Andreu D, Sulleiro E, Zarzuela F, Urtasun N, Pérez-Torras S, Pastor-Anglada M, Arce EM, Muñoz-Torrero D, Fernàndez-Busquets X. Leveraging the Aggregated Protein Dye YAT2150 for Malaria Chemotherapy. Pharmaceutics 2024; 16:1290. [PMID: 39458619 PMCID: PMC11514582 DOI: 10.3390/pharmaceutics16101290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: YAT2150 is a first-in-class antiplasmodial compound that has been recently proposed as a new interesting drug for malaria therapy. Methods/Results: The fluorescence of YAT2150 rapidly increases upon its entry into Plasmodium, a property that can be of use for the design of highly sensitive diagnostic approaches. YAT2150 blocks the in vitro development of the ookinete stage of Plasmodium and, when added to an infected blood meal, inhibits oocyst formation in the mosquito. Thus, the compound could possibly contribute to future transmission-blocking antimalarial strategies. Cell influx/efflux studies in Caco-2 cells suggest that YAT2150 is internalized by endocytosis and also through the OATP2B1 transporter, whereas its main export route would be via OSTα. YAT2150 has an overall favorable drug metabolism and pharmacokinetics profile, and its moderate cytotoxicity can be significantly reduced upon encapsulation in immunoliposomes, which leads to a dramatic increase in the drug selectivity index to values close to 1000. Although YAT2150 binds amyloid-forming peptides, its in vitro fluorescence emission is stronger upon association with peptides that form amorphous aggregates, suggesting that regions enriched in unstructured proteins are the preferential binding sites of the drug inside Plasmodium cells. The reduction of protein aggregation in the parasite after YAT2150 treatment, which has been suggested to be directly related to the drug's mode of action, is also observed following treatment with quinoline antimalarials like chloroquine and primaquine. Conclusions: Altogether, the data presented here indicate that YAT2150 can represent the spearhead of a new family of compounds for malaria diagnosis and therapy due to its presumed novel mode of action based on the interaction with functional protein aggregates in the pathogen.
Collapse
Affiliation(s)
- Claudia Camarero-Hoyos
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Doctoral School of Biotechnology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain
| | - Inés Bouzón-Arnáiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Yunuen Avalos-Padilla
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Antonino Nicolò Fallica
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Lucía Román-Álamo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Miriam Ramírez
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
| | - Emma Portabella
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Ona Cuspinera
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Daniela Currea-Ayala
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Marc Orozco-Quer
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Maria Ribera
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
| | - Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, FORTH, N. Plastira 100, 700 13 Heraklion, Greece; (I.S.-K.); (L.S.)
| | - Lefteris Spanos
- Institute of Molecular Biology and Biotechnology, FORTH, N. Plastira 100, 700 13 Heraklion, Greece; (I.S.-K.); (L.S.)
| | - Valentín Iglesias
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Clinical Research Centre, Medical University of Białystok, Kilińskiego 1, 15-369 Białystok, Poland
| | - Benigno Crespo
- Global Health Medicines R&D, GlaxoSmithKline (GSK), 28760 Tres Cantos, Spain; (B.C.); (S.V.)
| | - Sara Viera
- Global Health Medicines R&D, GlaxoSmithKline (GSK), 28760 Tres Cantos, Spain; (B.C.); (S.V.)
| | - David Andreu
- Department of Medicine and Life Sciences, Barcelona Biomedical Research Park, Pompeu Fabra University, Dr. Aiguader 88, 08003 Barcelona, Spain;
| | - Elena Sulleiro
- Microbiology Department, Vall d’Hebron University Hospital (VHUH), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (E.S.); (F.Z.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Carlos III Health Institute, 28029 Madrid, Spain
| | - Francesc Zarzuela
- Microbiology Department, Vall d’Hebron University Hospital (VHUH), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (E.S.); (F.Z.)
| | - Nerea Urtasun
- Molecular Pharmacology and Experimental Therapeutics (MPET), Department of Biochemistry and Molecular Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain; (N.U.); (S.P.-T.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Carlos III Health Institute, 28029 Madrid, Spain
- Institut de Recerca Hospital Sant Joan de Déu de Barcelona (IRSJD), Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
| | - Sandra Pérez-Torras
- Molecular Pharmacology and Experimental Therapeutics (MPET), Department of Biochemistry and Molecular Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain; (N.U.); (S.P.-T.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Carlos III Health Institute, 28029 Madrid, Spain
- Institut de Recerca Hospital Sant Joan de Déu de Barcelona (IRSJD), Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
| | - Marçal Pastor-Anglada
- Molecular Pharmacology and Experimental Therapeutics (MPET), Department of Biochemistry and Molecular Biology, University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain; (N.U.); (S.P.-T.); (M.P.-A.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Carlos III Health Institute, 28029 Madrid, Spain
- Institut de Recerca Hospital Sant Joan de Déu de Barcelona (IRSJD), Santa Rosa 39-57, 08950 Esplugues de Llobregat, Spain
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
| | - Elsa M. Arce
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
| | - Diego Muñoz-Torrero
- Institute of Biomedicine (IBUB), University of Barcelona, Av. Diagonal 643, 08028 Barcelona, Spain;
- Laboratory of Medicinal Chemistry (CSIC Associated Unit), Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII 27-31, 08028 Barcelona, Spain;
| | - Xavier Fernàndez-Busquets
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic-Universitat de Barcelona, Rosselló 149-153, 08036 Barcelona, Spain; (C.C.-H.); (I.B.-A.); (Y.A.-P.); (A.N.F.); (L.R.-Á.); (M.R.); (E.P.); (O.C.); (D.C.-A.); (M.O.-Q.); (M.R.); (V.I.)
- Nanomalaria Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 10-12, 08028 Barcelona, Spain
- Nanoscience and Nanotechnology Institute (IN2UB), University of Barcelona, Martí i Franquès 1, 08028 Barcelona, Spain
| |
Collapse
|
7
|
Cheng M, Ren L, Jia X, Wang J, Cong B. Understanding the action mechanisms of metformin in the gastrointestinal tract. Front Pharmacol 2024; 15:1347047. [PMID: 38617792 PMCID: PMC11010946 DOI: 10.3389/fphar.2024.1347047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/15/2024] [Indexed: 04/16/2024] Open
Abstract
Metformin is the initial medication recommended for the treatment of type 2 diabetes mellitus (T2DM). In addition to diabetes treatment, the function of metformin also can be anti-aging, antiviral, and anti-inflammatory. Nevertheless, further exploration is required to fully understand its mode of operation. Historically, the liver has been acknowledged as the main location where metformin reduces glucose levels, however, there is increasing evidence suggesting that the gastrointestinal tract also plays a significant role in its action. In the gastrointestinal tract, metformin effects glucose uptake and absorption, increases glucagon-like peptide-1 (GLP-1) secretion, alters the composition and structure of the gut microbiota, and modulates the immune response. However, the side effects of it cannot be ignored such as gastrointestinal distress in patients. This review outlines the impact of metformin on the digestive system and explores potential explanations for variations in metformin effectiveness and adverse effects like gastrointestinal discomfort.
Collapse
Affiliation(s)
- Meihui Cheng
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lili Ren
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xianxian Jia
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Pathogen Biology, Institute of Basic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Jianwei Wang
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Cong
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- College of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Perazza F, Leoni L, Colosimo S, Musio A, Bocedi G, D’Avino M, Agnelli G, Nicastri A, Rossetti C, Sacilotto F, Marchesini G, Petroni ML, Ravaioli F. Metformin and the Liver: Unlocking the Full Therapeutic Potential. Metabolites 2024; 14:186. [PMID: 38668314 PMCID: PMC11052067 DOI: 10.3390/metabo14040186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Metformin is a highly effective medication for managing type 2 diabetes mellitus. Recent studies have shown that it has significant therapeutic benefits in various organ systems, particularly the liver. Although the effects of metformin on metabolic dysfunction-associated steatotic liver disease and metabolic dysfunction-associated steatohepatitis are still being debated, it has positive effects on cirrhosis and anti-tumoral properties, which can help prevent the development of hepatocellular carcinoma. Furthermore, it has been proven to improve insulin resistance and dyslipidaemia, commonly associated with liver diseases. While more studies are needed to fully determine the safety and effectiveness of metformin use in liver diseases, the results are highly promising. Indeed, metformin has a terrific potential for extending its full therapeutic properties beyond its traditional use in managing diabetes.
Collapse
Affiliation(s)
- Federica Perazza
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Laura Leoni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Santo Colosimo
- Doctorate School of Nutrition Science, University of Milan, 20122 Milan, Italy;
| | | | - Giulia Bocedi
- U.O. Diabetologia, Ospedale C. Magati, Scandiano, 42019 Reggio Emilia, Italy;
| | - Michela D’Avino
- S.C. Endocrinologia Arcispedale Santa Maria Nuova, 42123 Reggio Emilia, Italy;
| | - Giulio Agnelli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Alba Nicastri
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Chiara Rossetti
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federica Sacilotto
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Giulio Marchesini
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Maria Letizia Petroni
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
| | - Federico Ravaioli
- Department of Medical and Surgical Sciences, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy; (F.P.); (L.L.); (G.A.); (A.N.); (C.R.); (F.S.); (G.M.); (M.L.P.)
- Division of Hepatobiliary and Immunoallergic Diseases, Department of Internal Medicine, IRCCS Azienda Ospedaliero, Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
9
|
Fan N, Du L, Guo T, Liu M, Chen X. Pharmacokinetic Interaction Between Imatinib and Metformin in Rats. Eur J Drug Metab Pharmacokinet 2024; 49:171-179. [PMID: 38141154 DOI: 10.1007/s13318-023-00869-x] [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] [Accepted: 11/12/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND AND OBJECTIVE Imatinib is primarily transported into the liver by organic cation transporter 1 (OCT1), organic anion transporting polypeptide 1B3 (OATP1B3), and novel organic cation transporter 2 (OCTN2), which is the first step in the metabolic and elimination of imatinib. Patients taking imatinib may concurrently take metformin, a substrate for OCT1. Drug-drug interactions (DDI) may occur between imatinib and metformin, affecting the clinical efficacy of imatinib. This experiment aimed to investigate the pharmacokinetic effects of metformin on imatinib and its active metabolism of N-desmethyl imatinib in rats. METHODS Twenty healthy Sprague-Dawley rats were selected and randomly divided into control and experimental groups (10 rats per group). The control group was orally administered imatinib (30 mg/kg) for 14 days, and the experimental group was orally co-administered imatinib (30 mg/kg) and metformin (200 mg/kg) for 14 days. The plasma concentrations of imatinib and N-desmethyl imatinib in rats were determined by ultra-performance liquid chromatography-mass spectrometry. Pharmacokinetic parameters were calculated by DAS2.0 software. RESULTS After single-dose co-administration of imatinib and metformin on day 1, the AUC0-24 (area under the plasma concentration-time curve) and Cmax (maximum concentration) of imatinib and the MRT (mean residence time) and Cmax of N-desmethyl imatinib in the experimental group were significantly decreased compared with the control group (P < 0.05). After multiple-dose co-administration of imatinib and metformin for 14 days, the AUC0-24 and Cmax of both imatinib and N-desmethyl imatinib were significantly decreased in the experimental group (P < 0.05). CONCLUSION With both single and multiple co-administration doses, metformin significantly changed the pharmacokinetic parameters of imatinib and N-desmethyl imatinib. The results suggest that care should be taken when metformin and imatinib are co-administered.
Collapse
Affiliation(s)
- Naling Fan
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of clinical Pharmacy, Health Road, Chang'an District, Shijiazhuang City, Hebei Province, People's Republic of China
| | - Liying Du
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of clinical Pharmacy, Health Road, Chang'an District, Shijiazhuang City, Hebei Province, People's Republic of China
| | - Teng Guo
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of clinical Pharmacy, Health Road, Chang'an District, Shijiazhuang City, Hebei Province, People's Republic of China
| | - Mingfeng Liu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of clinical Pharmacy, Health Road, Chang'an District, Shijiazhuang City, Hebei Province, People's Republic of China
| | - Xinran Chen
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Hebei Key Laboratory of clinical Pharmacy, Health Road, Chang'an District, Shijiazhuang City, Hebei Province, People's Republic of China.
| |
Collapse
|
10
|
Kang MJ, Kim MJ, Kim A, Koo TS, Lee KR, Chae YJ. Pharmacokinetic interactions of niclosamide in rats: Involvement of organic anion transporters 1 and 3 and organic cation transporter 2. Chem Biol Interact 2024; 390:110886. [PMID: 38280639 DOI: 10.1016/j.cbi.2024.110886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/30/2023] [Accepted: 01/19/2024] [Indexed: 01/29/2024]
Abstract
Niclosamide is an anthelmintic drug with a long history of use and is generally safe and well tolerated in humans. As the conventional dose of niclosamide results in a low but certain level in systemic circulation, drug interactions with concomitant drugs should be considered. We aimed to investigate the interaction between niclosamide and drug transporters, as such information is currently limited. Niclosamide inhibited the transport activity of OATP1B1, OATP1B3, OAT1, OAT3, and OCT2 in vitro. Among them, the inhibitory effects on OAT1, OAT3, and OCT2 were strong, with IC50 values of less than 1 μM. When 3 mg/kg of niclosamide was co-administered to rats, systemic exposure to furosemide (a substrate of OAT1/3) and metformin (a substrate of OCT2) increased, and the renal clearance (CLr) of the drugs significantly decreased. These results suggest that niclosamide inhibits renal transporters, OAT1/3 and OCT2, not only in vitro but also in vivo, resulting in increased systemic exposure to the substrates of the transporters by strongly blocking the urinary elimination pathway in rats. The findings of this study will support a meticulous understanding of the transporter-mediated drug interactions of niclosamide and consequently aid in effective and safe use of niclosamide.
Collapse
Affiliation(s)
- Min-Ji Kang
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea
| | - Min Ju Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Aeran Kim
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea
| | - Tae-Sung Koo
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea; Department of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju, 55338, Republic of Korea; Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju, 55338, Republic of Korea.
| |
Collapse
|
11
|
Ochi T, de Vos S, Touw D, Denig P, Feenstra T, Hak E. Tailoring Type II Diabetes Treatment: Investigating the Effect of 5-HTT Polymorphisms on HbA1c Levels after Metformin Initiation. J Diabetes Res 2024; 2024:7922486. [PMID: 38288388 PMCID: PMC10824573 DOI: 10.1155/2024/7922486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 11/10/2023] [Accepted: 12/19/2023] [Indexed: 01/31/2024] Open
Abstract
Aims To investigate the effect of serotonin transporter (5-HTT) polymorphisms on change in HbA1c levels six months after metformin initiation in type 2 diabetes patients. Materials and Methods Participants of PROVALID (PROspective cohort study in patients with type 2 diabetes mellitus for VALidation of biomarkers) within the GIANTT (Groningen Initiative to ANalyse Type 2 Diabetes Treatment) cohort who initiated metformin were genotyped for combined 5-HTTLPR/rs25531 (L∗L∗, L∗S∗, and S∗S∗) and 5-HTT VNTR (STin 2.12, 12/-, and 10/-) polymorphisms, respectively. Multiple linear regression was applied to determine the change in HbA1c level from baseline date to six months across 5-HTTLPR/VNTR genotype groups, adjusted for baseline HbA1c, age, gender, triglyceride level, low-density lipoprotein level, and serum creatinine. Results 157 participants were included, of which 56.2% were male. The average age was 59.3 ± 9.23 years, and the mean baseline HbA1c was 7.49% ± 1.21%. 5-HTTLPR was characterized in 46 patients as L∗L∗, 70 patients as L∗S∗, and 41 patients as S∗S∗ genotypes. No significant association was found between 5-HTTLPR and 5-HTT VNTR genotypes and change in HbA1c after adjustments. Conclusions 5-HTT polymorphisms did not affect HbA1c levels six months after the start of metformin. Further long-term studies in large samples would be relevant to determine which polymorphisms can explain the variation in response to metformin treatment.
Collapse
Affiliation(s)
- Taichi Ochi
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
| | - Stijn de Vos
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
| | - Daan Touw
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pharmacokinetics, Toxicology and Targeting, Groningen, Netherlands
| | - Petra Denig
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Talitha Feenstra
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
- Dutch National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Eelko Hak
- Groningen Research Institute of Pharmacy, PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, Netherlands
| |
Collapse
|
12
|
Honan LE, Fraser-Spears R, Daws LC. Organic cation transporters in psychiatric and substance use disorders. Pharmacol Ther 2024; 253:108574. [PMID: 38072333 PMCID: PMC11052553 DOI: 10.1016/j.pharmthera.2023.108574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/01/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Psychiatric and substance use disorders inflict major public health burdens worldwide. Their widespread burden is compounded by a dearth of effective treatments, underscoring a dire need to uncover novel therapeutic targets. In this review, we summarize the literature implicating organic cation transporters (OCTs), including three subtypes of OCTs (OCT1, OCT2, and OCT3) and the plasma membrane monoamine transporter (PMAT), in the neurobiology of psychiatric and substance use disorders with an emphasis on mood and anxiety disorders, alcohol use disorder, and psychostimulant use disorder. OCTs transport monoamines with a low affinity but high capacity, situating them to play a central role in regulating monoamine homeostasis. Preclinical evidence discussed here suggests that OCTs may serve as promising targets for treatment of psychiatric and substance use disorders and encourage future research into their therapeutic potential.
Collapse
Affiliation(s)
- Lauren E Honan
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA
| | - Rheaclare Fraser-Spears
- University of the Incarnate Word, Feik School of Pharmacy, Department of Pharmaceutical Sciences, USA
| | - Lynette C Daws
- The University of Texas Health Science Center at San Antonio, Department of Cellular & Integrative Physiology, USA; The University of Texas Health Science Center at San Antonio, Department of Pharmacology, USA.
| |
Collapse
|
13
|
Oyanna VO, Garcia-Torres KY, Bechtold BJ, Lynch KD, Call MR, Horváth M, Manwill PK, Graf TN, Cech NB, Oberlies NH, Paine MF, Clarke JD. Goldenseal-Mediated Inhibition of Intestinal Uptake Transporters Decreases Metformin Systemic Exposure in Mice. Drug Metab Dispos 2023; 51:1483-1489. [PMID: 37562957 PMCID: PMC10586506 DOI: 10.1124/dmd.123.001360] [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: 04/11/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
Goldenseal is a perennial plant native to eastern North America. A recent clinical study reported goldenseal decreased metformin Cmax and area under the blood concentration versus time curve (AUC) by 27% and 23%, respectively, but half-life and renal clearance were unchanged. These observations suggested goldenseal altered processes involved in metformin absorption. The underlying mechanism(s) remain(s) unknown. One mechanism for the decreased metformin systemic exposure is inhibition by goldenseal of intestinal uptake transporters involved in metformin absorption. Goldenseal extract and three goldenseal alkaloids (berberine, (-)-β-hydrastine, hydrastinine) were tested as inhibitors of organic cation transporter (OCT) 3, plasma membrane monoamine transporter (PMAT), and thiamine transporter (THTR) 2 using human embryonic kidney 293 cells overexpressing each transporter. The goldenseal extract, normalized to berberine content, was the strongest inhibitor of each transporter (IC50: 4.9, 13.1, and 5.8 μM for OCT3, PMAT, and THTR2, respectively). A pharmacokinetic study in mice compared the effects of berberine, (-)-β-hydrastine, goldenseal extract, and imatinib (OCT inhibitor) on orally administered metformin. Goldenseal extract and imatinib significantly decreased metformin Cmax by 31% and 25%, respectively, and had no effect on half-life. Berberine and (-)-β-hydrastine had no effect on metformin pharmacokinetics, indicating neither alkaloid alone precipitated the interaction in vivo. A follow-up murine study involving intravenous metformin and oral inhibitors examined the contributions of basolateral enteric/hepatic uptake transporters to the goldenseal-metformin interaction. Goldenseal extract and imatinib had no effect on metformin AUC and half-life, suggesting lack of inhibition of basolateral enteric/hepatic uptake transporters. Results may have implications for patients taking goldenseal with drugs that are substrates for OCT3 and THTR2. SIGNIFICANCE STATEMENT: Goldenseal is used to self-treat respiratory infections and digestive disorders. We investigated potential mechanisms for the clinical pharmacokinetic interaction observed between goldenseal and metformin, specifically inhibition by goldenseal of intestinal uptake transporters (OCT3, PMAT, THTR2) involved in metformin absorption. Goldenseal extract inhibited all three transporters in vitro and decreased metformin systemic exposure in mice. These data may have broader implications for patients co-consuming goldenseal with other drugs that are substrates for these transporters.
Collapse
Affiliation(s)
- Victoria O Oyanna
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Kenisha Y Garcia-Torres
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Baron J Bechtold
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Katherine D Lynch
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - M Ridge Call
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Miklós Horváth
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Preston K Manwill
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Tyler N Graf
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Nadja B Cech
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Nicholas H Oberlies
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - Mary F Paine
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| | - John D Clarke
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington (V.O.O., K.Y.G.-T., B.J.B., K.D.L., M.R.C., M.F.P., J.D.C.); Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, North Carolina (P.K.M., T.N.G., N.B.C., N.H.O.); SOLVO Biotechnology, Szeged, Hungary (M.H.); and Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington (N.B.C., N.H.O., M.F.P., J.D.C.)
| |
Collapse
|
14
|
Alam S, Doherty E, Ortega-Prieto P, Arizanova J, Fets L. Membrane transporters in cell physiology, cancer metabolism and drug response. Dis Model Mech 2023; 16:dmm050404. [PMID: 38037877 PMCID: PMC10695176 DOI: 10.1242/dmm.050404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Abstract
By controlling the passage of small molecules across lipid bilayers, membrane transporters influence not only the uptake and efflux of nutrients, but also the metabolic state of the cell. With more than 450 members, the Solute Carriers (SLCs) are the largest transporter super-family, clustering into families with different substrate specificities and regulatory properties. Cells of different types are, therefore, able to tailor their transporter expression signatures depending on their metabolic requirements, and the physiological importance of these proteins is illustrated by their mis-regulation in a number of disease states. In cancer, transporter expression is heterogeneous, and the SLC family has been shown to facilitate the accumulation of biomass, influence redox homeostasis, and also mediate metabolic crosstalk with other cell types within the tumour microenvironment. This Review explores the roles of membrane transporters in physiological and malignant settings, and how these roles can affect drug response, through either indirect modulation of sensitivity or the direct transport of small-molecule therapeutic compounds into cells.
Collapse
Affiliation(s)
- Sara Alam
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Emily Doherty
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Paula Ortega-Prieto
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Julia Arizanova
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| | - Louise Fets
- Drug Transport and Tumour Metabolism Lab, MRC Laboratory of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
15
|
Lofthouse EM, Cleal J, Lewis RM, Sengers BG. Computational Modelling of Paracellular Diffusion and OCT3 Mediated Transport of Metformin in the Perfused Human Placenta. J Pharm Sci 2023; 112:2570-2580. [PMID: 37211316 DOI: 10.1016/j.xphs.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/23/2023]
Abstract
Metformin is an antidiabetic drug, increasingly prescribed in pregnancy and has been shown to cross the human placenta. The mechanisms underlying placental metformin transfer remain unclear. This study investigated the roles of drug transporters and paracellular diffusion in the bidirectional transfer of metformin across the human placental syncytiotrophoblast using placental perfusion experiments and computational modelling. 14C-metformin transfer was observed in the maternal to fetal and fetal to maternal directions and was not competitively inhibited by 5 mM unlabelled metformin. Computational modelling of the data was consistent with overall placental transfer via paracellular diffusion. Interestingly, the model also predicted a transient peak in fetal 14C-metformin release due to trans-stimulation of OCT3 by unlabelled metformin at the basal membrane. To test this hypothesis a second experiment was designed. OCT3 substrates (5 mM metformin, 5 mM verapamil and 10 mM decynium-22) added to the fetal artery trans-stimulated release of 14C-metformin from the placenta into the fetal circulation, while 5 mM corticosterone did not. This study demonstrated activity of OCT3 transporters on the basal membrane of the human syncytiotrophoblast. However, we did not detect a contribution of either OCT3 or apical membrane transporters to overall materno-fetal transfer, which could be represented adequately by paracellular diffusion in our system.
Collapse
Affiliation(s)
- Emma M Lofthouse
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | - Jane Cleal
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | - Rohan M Lewis
- Faculty of Medicine, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK
| | - Bram G Sengers
- Faculty of Engineering and Physical Sciences, University of Southampton, UK; Institute for Life Sciences, University of Southampton, UK.
| |
Collapse
|
16
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
17
|
Sharma S, Zhang Y, Akter KA, Nozohouri S, Archie SR, Patel D, Villalba H, Abbruscato T. Permeability of Metformin across an In Vitro Blood-Brain Barrier Model during Normoxia and Oxygen-Glucose Deprivation Conditions: Role of Organic Cation Transporters (Octs). Pharmaceutics 2023; 15:1357. [PMID: 37242599 PMCID: PMC10220878 DOI: 10.3390/pharmaceutics15051357] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Our lab previously established that metformin, a first-line type two diabetes treatment, activates the Nrf2 pathway and improves post-stroke recovery. Metformin's brain permeability value and potential interaction with blood-brain barrier (BBB) uptake and efflux transporters are currently unknown. Metformin has been shown to be a substrate of organic cationic transporters (Octs) in the liver and kidneys. Brain endothelial cells at the BBB have been shown to express Octs; thus, we hypothesize that metformin uses Octs for its transport across the BBB. We used a co-culture model of brain endothelial cells and primary astrocytes as an in vitro BBB model to conduct permeability studies during normoxia and hypoxia using oxygen-glucose deprivation (OGD) conditions. Metformin was quantified using a highly sensitive LC-MS/MS method. We further checked Octs protein expression using Western blot analysis. Lastly, we completed a plasma glycoprotein (P-GP) efflux assay. Our results showed that metformin is a highly permeable molecule, uses Oct1 for its transport, and does not interact with P-GP. During OGD, we found alterations in Oct1 expression and increased permeability for metformin. Additionally, we showed that selective transport is a key determinant of metformin's permeability during OGD, thus, providing a novel target for improving ischemic drug delivery.
Collapse
Affiliation(s)
- Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Yong Zhang
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Khondker Ayesha Akter
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Sabrina Rahman Archie
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Dhavalkumar Patel
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Heidi Villalba
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Thomas Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
- Center for Blood-Brain Barrier Research, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| |
Collapse
|
18
|
Damanhouri ZA, Alkreathy HM, Alharbi FA, Abualhamail H, Ahmad MS. A Review of the Impact of Pharmacogenetics and Metabolomics on the Efficacy of Metformin in Type 2 Diabetes. Int J Med Sci 2023; 20:142-150. [PMID: 36619226 PMCID: PMC9812811 DOI: 10.7150/ijms.77206] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 12/02/2022] [Indexed: 01/06/2023] Open
Abstract
Metformin is the most often prescribed drug for people with type 2 diabetes (T2D). More than 120 million patients with T2D use metformin worldwide. However, monotherapy fails to achieve glycemic control in a third of the treated patients. Genetics contribute to some of the inter-individual variations in glycemic response to metformin. Numerous pharmacogenetic studies have demonstrated that variations in genes related to pharmacokinetics and pharmacodynamics of metformin's encoding transporters are mainly associated with metformin response. The goal of this review is to evaluate the current state of metformin pharmacogenetics and metabolomics research, discuss the clinical and scientific issues that need to be resolved in order to increase our knowledge of patient response variability to metformin, and how to improve patient outcomes. Metformin's hydrophilic nature and absorption as well as its action mechanism and effectiveness on T2D initiation are discussed. The impacts of variations associated with various genes are analysed to identify and evaluate the effect of genetic polymorphisms on the therapeutic activity of metformin. The metabolic pattern of T2D and metformin is also indicated. This is to emphasise that studies of pharmacogenetics and metabolomics could expand our knowledge of metformin response in T2D.
Collapse
Affiliation(s)
- Zoheir A Damanhouri
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda M Alkreathy
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fawaz A Alharbi
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haneen Abualhamail
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad S Ahmad
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
19
|
Li H, Wang X, Wang Y, Zhang M, Hong F, Wang H, Cui A, Zhao J, Ji W, Chen YG. Cross-species single-cell transcriptomic analysis reveals divergence of cell composition and functions in mammalian ileum epithelium. CELL REGENERATION 2022; 11:19. [PMID: 35511361 PMCID: PMC9072607 DOI: 10.1186/s13619-022-00118-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022]
Abstract
AbstractAnimal models are widely used for biomedical studies and drug evaluation. The small intestine plays key roles in nutrient absorption, hormone secretion, microbiota defense and drug absorption and metabolism. Although the intestinal structure of mammals is conserved, the differences on epithelial cell composition, functional assignments and drug absorption among mammals are largely unknown. Here, cross-species analysis of single-cell transcriptomic atlas of the ileum epithelium from mouse, rat, pig, macaque and human reveals the conserved and differential cell types and functions among species, identifies a new CA7+ cell type in pig, macaque and human ileum, uncovers the distinct expression pattern in enterocytes, enteroendocrine cells and Paneth cells, and defines the conserved and species-specific intestinal stem cell signature genes. The examination of drug absorption across species suggests that drug metabolism in mouse ileum is closer to human while drug transport in macaque ileum is more similar to human. Together, our data provide the comprehensive information about cell composition and functional assignments in five species, and offer the valuable guidance for animal model selection and drug testing.
Collapse
|
20
|
ÇİFTÇİOĞLU M. Shouldn't Stage 4 And 5 Chronic Kidney Disease Patients Use Metformin? KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1181458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Metformin is the first place anti-diabetic agent recommended with life style changes in many guidelines for the treatment of patients with type 2 diabetes mellitus (DM). The mechanism of effect of the drug is to increase insulin sensitivity in peripheral tissue and reduce glucose secretion from the liver. Metformin is a low cost, effective and safe drug. Although its frequent side effects are gastrointestinal side effects and the most feared side effect is lactic acidosis. Due to this side effect, its use is limited in many guidelines in patients with chronic kidney disease (CKD). In this article, we examined the use of metformin in all stages of CKD. We investigated the incidence of metformin-associated lactic acidosis (MALA). Shouldn't stage 4 and 5 chronic kidney disease patients use metformin? We sought an answer to question. As a result, we decided that side effects like MALA are extremely rare. We observed that these side effects occur mostly in the presence of diseases in which tissue perfusion is impaired such as infections, serious cardiovascular events, and hypotension. We came to the conclusion that metformin should be used in patients with stage 4 and 5 CKD patients, without much fear, considering the profit and loss relationship.
Collapse
|
21
|
Li S, Xu B, Fan S, Kang B, Deng L, Chen D, Yang B, Tang F, He Z, Xue Y, Zhou JC. Effects of single-nucleotide polymorphism on the pharmacokinetics and pharmacodynamics of metformin. Expert Rev Clin Pharmacol 2022; 15:1107-1117. [PMID: 36065506 DOI: 10.1080/17512433.2022.2118714] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Metformin has been recognized as the first-choice drug for type 2 diabetes mellitus (T2DM). The potency of metformin in the treatment of type 2 diabetes has always been in the spotlight and shown significant individual differences. Based on previous studies, the efficacy of metformin is related to the single-nucleotide polymorphisms of transporter genes carried by patients, amongst which a variety of gene polymorphisms of transporter and target protein genes affect the effectiveness and adverse repercussion of metformin. AREAS COVERED Here, we reviewed the current knowledge about gene polymorphisms impacting metformin efficacy based on transporter and drug target proteins. EXPERT OPINION The reason for the difference in clinical drug potency of metformin can be attributed to the gene polymorphism of drug transporters and drug target proteins in the human body. Substantial evidence shows that genetic polymorphisms in transporters such as organic cation transporter 1 (OCT1) and organic cation transporter 2 (OCT2) affect the glucose-lowering effectiveness of metformin. However, optimization of individualized dosing regimens of metformin is necessary to clarify the role of several polymorphisms.
Collapse
Affiliation(s)
- Shaoqian Li
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Xu
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Shangzhi Fan
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Kang
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lijing Deng
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Danjun Chen
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Bo Yang
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Fan Tang
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Zunbo He
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Department of Anesthesiology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yong Xue
- The Second Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jie-Can Zhou
- The First Affiliated Hospital, Clinical Pharmacology Research Center, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Hengyang Key Laboratory of Clinical Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China.,The First Affiliated Hospital, Pharmacy Department, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
22
|
The role of MicroRNA networks in tissue-specific direct and indirect effects of metformin and its application. Biomed Pharmacother 2022; 151:113130. [PMID: 35598373 DOI: 10.1016/j.biopha.2022.113130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/06/2022] [Accepted: 05/13/2022] [Indexed: 11/20/2022] Open
Abstract
Metformin is a first-line oral antidiabetic agent that results in clear benefits in relation to glucose metabolism and diabetes-related complications. The specific regulatory details and mechanisms underlying these benefits are still unclear and require further investigation. There is recent mounting evidence that metformin has pleiotropic effects on the target tissue development in metabolic organs, including adipose tissue, the gastrointestinal tract and the liver. The mechanism of actions of metformin are divided into direct effects on target tissues and indirect effects via non-targeted tissues. MicroRNAs (miRNAs) are a class of endogenous, noncoding, negative gene regulators that have emerged as important regulators of a number of diseases, including type 2 diabetes mellitus (T2DM). Metformin is involved in many aspects of miRNA regulation, and metformin treatment in T2DM should be associated with other miRNA targets. A large number of miRNAs regulation by metformin in target tissues with either direct or indirect effects has gradually been revealed in the context of numerous diseases and has gradually received increasing attention. This paper thoroughly reviews the current knowledge about the role of miRNA networks in the tissue-specific direct and indirect effects of metformin. Furthermore, this knowledge provides a novel theoretical basis and suggests therapeutic targets for the clinical treatment of metformin and miRNA regulators in the prevention and treatment of cancer, cardiovascular disorders, diabetes and its complications.
Collapse
|
23
|
Kurosawa K, Noguchi S, Nishimura T, Tomi M, Chiba K. Transplacental Pharmacokinetic Model of Digoxin Based on Ex Vivo Human Placental Perfusion Study. Drug Metab Dispos 2022; 50:287-298. [PMID: 34903589 DOI: 10.1124/dmd.121.000648] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/10/2021] [Indexed: 11/22/2022] Open
Abstract
Digoxin is used as first-line therapy to treat fetal supraventricular tachycardia; however, because of the narrow therapeutic window, it is essential to estimate digoxin exposure in the fetus. The data from ex vivo human placental perfusion study are used to predict in vivo fetal exposure noninvasively, but the ex vivo fetal-to-maternal concentration (F:M) ratios observed in digoxin perfusion studies were much lower than those in vivo. In the present study, we developed a human transplacental pharmacokinetic model of digoxin using previously reported ex vivo human placental perfusion data. The model consists of maternal intervillous, fetal capillary, non-perfused tissue, and syncytiotrophoblast compartments, with multidrug resistance protein (MDR) 1 and influx transporter at the microvillous membrane (MVM) and influx and efflux transporters at the basal plasma membrane (BM). The model-predicted F:M ratio was 0.66, which is consistent with the mean in vivo value of 0.77 (95% confidence interval: 0.64-0.91). The time to achieve the steady state from the ex vivo perfusion study was estimated as 1,500 minutes, which is considerably longer than the reported ex vivo experimental durations, and this difference is considered to account for the inconsistency between ex vivo and in vivo F:M ratios. Reported digoxin concentrations in a drug-drug interaction study with MDR1 inhibitors quinidine and verapamil were consistent with the profiles simulated by our model incorporating inhibition of efflux transporter at the BM in addition to MVM. Our modeling and simulation approach should be a powerful tool to predict fetal exposure and DDIs in human placenta. SIGNIFICANCE STATEMENT: We developed a human transplacental pharmacokinetic model of digoxin based on ex vivo human placental perfusion studies in order to resolve inconsistencies between reported ex vivo and in vivo fetal-to-maternal concentration ratios. The model successfully predicted the in vivo fetal exposure to digoxin and the drug-drug interactions of digoxin and P-glycoprotein/multidrug resistance protein 1 inhibitors in human placenta.
Collapse
Affiliation(s)
- Ken Kurosawa
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Saki Noguchi
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Tomohiro Nishimura
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Masatoshi Tomi
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Koji Chiba
- Department of Japan-Clinical Pharmacology and Pharmacometrics, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| |
Collapse
|
24
|
Shirasaka Y, Seki M, Hatakeyama M, Kurokawa Y, Uchiyama H, Takemura M, Yasugi Y, Kishimoto H, Tamai I, Wang J, Inoue K. Multiple Transport Mechanisms Involved in the Intestinal Absorption of Metformin: Impact on the Nonlinear Absorption Kinetics. J Pharm Sci 2022; 111:1531-1541. [DOI: 10.1016/j.xphs.2022.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/07/2022] [Accepted: 01/07/2022] [Indexed: 01/11/2023]
|
25
|
Hanke N, Türk D, Selzer D, Ishiguro N, Ebner T, Wiebe S, Müller F, Stopfer P, Nock V, Lehr T. A Comprehensive Whole-Body Physiologically Based Pharmacokinetic Drug-Drug-Gene Interaction Model of Metformin and Cimetidine in Healthy Adults and Renally Impaired Individuals. Clin Pharmacokinet 2021; 59:1419-1431. [PMID: 32449077 PMCID: PMC7658088 DOI: 10.1007/s40262-020-00896-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Background Metformin is a widely prescribed antidiabetic BCS Class III drug (low permeability) that depends on active transport for its absorption and disposition. It is recommended by the US Food and Drug Administration as a clinical substrate of organic cation transporter 2/multidrug and toxin extrusion protein for drug–drug interaction studies. Cimetidine is a potent organic cation transporter 2/multidrug and toxin extrusion protein inhibitor. Objective The objective of this study was to provide mechanistic whole-body physiologically based pharmacokinetic models of metformin and cimetidine, built and evaluated to describe the metformin-SLC22A2 808G>T drug–gene interaction, the cimetidine-metformin drug–drug interaction, and the impact of renal impairment on metformin exposure. Methods Physiologically based pharmacokinetic models were developed in PK-Sim® (version 8.0). Thirty-nine clinical studies (dosing range 0.001–2550 mg), providing metformin plasma and urine data, positron emission tomography measurements of tissue concentrations, studies in organic cation transporter 2 polymorphic volunteers, drug–drug interaction studies with cimetidine, and data from patients in different stages of chronic kidney disease, were used to develop the metformin model. Twenty-seven clinical studies (dosing range 100–800 mg), reporting cimetidine plasma and urine concentrations, were used for the cimetidine model development. Results The established physiologically based pharmacokinetic models adequately describe the available clinical data, including the investigated drug–gene interaction, drug–drug interaction, and drug–drug–gene interaction studies, as well as the metformin exposure during renal impairment. All modeled drug–drug interaction area under the curve and maximum concentration ratios are within 1.5-fold of the observed ratios. The clinical data of renally impaired patients shows the expected increase in metformin exposure with declining kidney function, but also indicates counter-regulatory mechanisms in severe renal disease; these mechanisms were implemented into the model based on findings in preclinical species. Conclusions Whole-body physiologically based pharmacokinetic models of metformin and cimetidine were built and qualified for the prediction of metformin pharmacokinetics during drug–gene interaction, drug–drug interaction, and different stages of renal disease. The model files will be freely available in the Open Systems Pharmacology model repository. Current guidelines for metformin treatment of renally impaired patients should be reviewed to avoid overdosing in CKD3 and to allow metformin therapy of CKD4 patients. Electronic supplementary material The online version of this article (10.1007/s40262-020-00896-w) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nina Hanke
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | - Denise Türk
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | - Dominik Selzer
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany
| | - Naoki Ishiguro
- Kobe Pharma Research Institute, Nippon Boehringer Ingelheim Co. Ltd., Kobe, Japan
| | - Thomas Ebner
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Sabrina Wiebe
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.,Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Fabian Müller
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Stopfer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Valerie Nock
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123, Saarbrücken, Germany.
| |
Collapse
|
26
|
Oh S, Cho Y, Chang M, Park S, Kwon H. Metformin Decreases 2-HG Production through the MYC-PHGDH Pathway in Suppressing Breast Cancer Cell Proliferation. Metabolites 2021; 11:metabo11080480. [PMID: 34436421 PMCID: PMC8402004 DOI: 10.3390/metabo11080480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022] Open
Abstract
The biguanide drug metformin has been widely used for the treatment of type 2 diabetes, and there is evidence supporting the anticancer effect of metformin despite some controversy. Here, we report the growth inhibitory activity of metformin in the breast cancer (MCF-7) cells, both in vitro and in vivo, and the associated metabolic changes. In particular, a decrease in a well-known oncometabolite 2-hydroxyglutarate (2-HG) was discovered by a metabolomics approach. The decrease in 2-HG by metformin was accompanied by the reduction in histone methylation, consistent with the known tumorigenic mechanism of 2-HG. The relevance of 2-HG inhibition in breast cancer was also supported by a higher level of 2-HG in human breast cancer tissues. Genetic knockdown of PHGDH identified the PHGDH pathway as the producer of 2-HG in the MCF-7 cells that do not carry isocitrate dehydrogenase 1 and 2 (IDH1/IDH2) mutations, the conventional producer of 2-HG. We also showed that metformin’s inhibitory effect on the PHGDH-2HG axis may occur through the regulation of the AMPK-MYC pathway. Overall, our results provide an explanation for the coherent pathway from complex I inhibition to epigenetic changes for metformin’s anticancer effect.
Collapse
Affiliation(s)
- Sehyun Oh
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
| | - Youngup Cho
- Department of Surgery, College of Medicine, Inha University, Inhang-Ro 27, Chung-gu, Incheon 22332, Korea;
| | - Minsun Chang
- Department of Biological Sciences, College of Science, Sookmyung Women’s University, 100, Cheongpa-ro 47-gil, Yongsan-gu, Seoul 140-742, Korea
- Correspondence: (M.C.); (S.P.); (H.K.)
| | - Sunghyouk Park
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
- Correspondence: (M.C.); (S.P.); (H.K.)
| | - Hyuknam Kwon
- Natural Product Research Institute, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
- Department of Biological and Environmental Sciences, University of Helsinki, 00160 Helsinki, Finland
- Correspondence: (M.C.); (S.P.); (H.K.)
| |
Collapse
|
27
|
Xiao D, Liu JY, Zhang SM, Liu RR, Yin JY, Han XY, Li X, Zhang W, Chen XP, Zhou HH, Ji LN, Liu ZQ. A Two-Stage Study Identifies Two Novel Polymorphisms in PRKAG2 Affecting Metformin Response in Chinese Type 2 Diabetes Patients. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2021; 14:745-755. [PMID: 34188521 PMCID: PMC8236263 DOI: 10.2147/pgpm.s305020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/25/2021] [Indexed: 12/29/2022]
Abstract
Objective Individual differences in glycemic response to metformin in antidiabetic treatment exist widely. Although some associated genetic variations have been discovered, they still cannot accurately predict metformin response. In the current study, we set out to investigate novel genetic variants affecting metformin response in Chinese type 2 diabetes (T2D) patients. Methods A two-stage study enrolled 500 T2D patients who received metformin, glibenclamide or a combination of both were recruited from 2009 to 2012 in China. Change of HbA1c, adjusted by clinical covariates, was used to evaluate glycemic response to metformin. Selected single nucleotide polymorphisms (SNPs) were genotyped using the Infinium iSelect and/or Illumina GoldenGate genotyping platform. A linear regression model was used to evaluate the association between SNPs and response. Results A total of 3739 SNPs were screened in Stage 1, of which 50 were associated with drug response. Except for one genetic variant preferred to affect glibenclamide, the remaining SNPs were subsequently verified in Stage 2, and two SNPs were successfully validated. These were PRKAG2 rs2727528 (discovery group: β=−0.212, P=0.046; validation group: β=−0.269, P=0.028) and PRKAG2 rs1105842 (discovery group: β=0.205, P=0.048; validation group: β=0.273, P=0.025). C allele carriers of rs2727528 and C allele carriers of rs1105842 would have a larger difference of HbA1c level when using metformin. Conclusion Two variants rs2727528 and rs1105842 in PRKAG2, encoding γ2 subunit of AMP-activated protein kinase (AMPK), were found to be associated with metformin response in Chinese T2D patients. These findings may provide some novel information for personalized pharmacotherapy of metformin in China.
Collapse
Affiliation(s)
- Di Xiao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Department of pharmacy, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Jun-Yan Liu
- Department of orthopaedics, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Si-Min Zhang
- Department of Endocrinology and Metabolism, The People's Hospital of Peking University, Beijing, People's Republic of China
| | - Rang-Ru Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Key Laboratory of Tropical Diseases and Translational Medicine of the Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical College, Haikou, People's Republic of China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China
| | - Xue-Yao Han
- Department of Endocrinology and Metabolism, The People's Hospital of Peking University, Beijing, People's Republic of China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Xiao-Ping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Li-Nong Ji
- Department of Endocrinology and Metabolism, The People's Hospital of Peking University, Beijing, People's Republic of China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
28
|
Baye AM, Fanta TG, Siddiqui MK, Dawed AY. The Genetics of Adverse Drug Outcomes in Type 2 Diabetes: A Systematic Review. Front Genet 2021; 12:675053. [PMID: 34194474 PMCID: PMC8236944 DOI: 10.3389/fgene.2021.675053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Adverse drug reactions (ADR) are a major clinical problem accounting for significant hospital admission rates, morbidity, mortality, and health care costs. One-third of people with diabetes experience at least one ADR. However, there is notable interindividual heterogeneity resulting in patient harm and unnecessary medical costs. Genomics is at the forefront of research to understand interindividual variability, and there are many genotype-drug response associations in diabetes with inconsistent findings. Here, we conducted a systematic review to comprehensively examine and synthesize the effect of genetic polymorphisms on the incidence of ADRs of oral glucose-lowering drugs in people with type 2 diabetes. Methods: A literature search was made to identify articles that included specific results of research on genetic polymorphism and adverse effects associated with oral glucose-lowering drugs. The electronic search was carried out on 3rd October 2020, through Cochrane Library, PubMed, and Web of Science using keywords and MeSH terms. Result: Eighteen articles consisting of 10, 383 subjects were included in this review. Carriers of reduced-function alleles of organic cation transporter 1 (OCT 1, encoded by SLC22A1) or reduced expression alleles of plasma membrane monoamine transporter (PMAT, encoded by SLC29A4) or serotonin transporter (SERT, encoded by SLC6A4) were associated with increased incidence of metformin-related gastrointestinal (GI) adverse effects. These effects were shown to exacerbate by concomitant treatment with gut transporter inhibiting drugs. The CYP2C9 alleles, *2 (rs1799853C>T) and *3 (rs1057910A>C) that are predictive of low enzyme activity were more common in subjects who experienced hypoglycemia after treatment with sulfonylureas. However, there was no significant association between sulfonylurea-related hypoglycemia and genetic variants in the ATP-binding cassette transporter sub-family C member 8 (ABCC8)/Potassium Inwardly Rectifying Channel Subfamily J Member 11 (KCNJ11). Compared to the wild type, the low enzyme activity C allele at CYP2C8*3 (rs1057910A>C) was associated with less weight gain whereas the C allele at rs6123045 in the NFATC2 gene was significantly associated with edema from rosiglitazone treatment. Conclusion: In spite of limited studies investigating genetics and ADR in diabetes, some convincing results are emerging. Genetic variants in genes encoding drug transporters and metabolizing enzymes are implicated in metformin-related GI adverse effects, and sulfonylurea-induced hypoglycemia, respectively. Further studies to investigate newer antidiabetic drugs such as DPP-4i, GLP-1RA, and SGLT2i are warranted. In addition, pharmacogenetic studies that account for race and ethnic differences are required.
Collapse
Affiliation(s)
- Assefa M Baye
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Teferi G Fanta
- Department of Pharmacology and Clinical Pharmacy, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Moneeza K Siddiqui
- Division of Population Health and Genomics, Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| | - Adem Y Dawed
- Division of Population Health and Genomics, Ninewells Hospital and School of Medicine, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
29
|
The Hormetic Effect of Metformin: "Less Is More"? Int J Mol Sci 2021; 22:ijms22126297. [PMID: 34208371 PMCID: PMC8231127 DOI: 10.3390/ijms22126297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/06/2021] [Accepted: 06/10/2021] [Indexed: 02/06/2023] Open
Abstract
Metformin (MTF) is the first-line therapy for type 2 diabetes (T2DM). The euglycemic effect of MTF is due to the inhibition of hepatic glucose production. Literature reports that the principal molecular mechanism of MTF is the activation of 5′-AMP-activated protein kinase (AMPK) due to the decrement of ATP intracellular content consequent to the inhibition of Complex I, although this effect is obtained only at millimolar concentrations. Conversely, micromolar MTF seems to activate the mitochondrial electron transport chain, increasing ATP production and limiting oxidative stress. This evidence sustains the idea that MTF exerts a hormetic effect based on its concentration in the target tissue. Therefore, in this review we describe the effects of MTF on T2DM on the principal target organs, such as liver, gut, adipose tissue, endothelium, heart, and skeletal muscle. In particular, data indicate that all organs, except the gut, accumulate MTF in the micromolar range when administered in therapeutic doses, unmasking molecular mechanisms that do not depend on Complex I inhibition.
Collapse
|
30
|
Interaction between Metformin, Folate and Vitamin B 12 and the Potential Impact on Fetal Growth and Long-Term Metabolic Health in Diabetic Pregnancies. Int J Mol Sci 2021; 22:ijms22115759. [PMID: 34071182 PMCID: PMC8198407 DOI: 10.3390/ijms22115759] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022] Open
Abstract
Metformin is the first-line treatment for many people with type 2 diabetes mellitus (T2DM) and gestational diabetes mellitus (GDM) to maintain glycaemic control. Recent evidence suggests metformin can cross the placenta during pregnancy, thereby exposing the fetus to high concentrations of metformin and potentially restricting placental and fetal growth. Offspring exposed to metformin during gestation are at increased risk of being born small for gestational age (SGA) and show signs of ‘catch up’ growth and obesity during childhood which increases their risk of future cardiometabolic diseases. The mechanisms by which metformin impacts on the fetal growth and long-term health of the offspring remain to be established. Metformin is associated with maternal vitamin B12 deficiency and antifolate like activity. Vitamin B12 and folate balance is vital for one carbon metabolism, which is essential for DNA methylation and purine/pyrimidine synthesis of nucleic acids. Folate:vitamin B12 imbalance induced by metformin may lead to genomic instability and aberrant gene expression, thus promoting fetal programming. Mitochondrial aerobic respiration may also be affected, thereby inhibiting placental and fetal growth, and suppressing mammalian target of rapamycin (mTOR) activity for cellular nutrient transport. Vitamin supplementation, before or during metformin treatment in pregnancy, could be a promising strategy to improve maternal vitamin B12 and folate levels and reduce the incidence of SGA births and childhood obesity. Heterogeneous diagnostic and screening criteria for GDM and the transient nature of nutrient biomarkers have led to inconsistencies in clinical study designs to investigate the effects of metformin on folate:vitamin B12 balance and child development. As rates of diabetes in pregnancy continue to escalate, more women are likely to be prescribed metformin; thus, it is of paramount importance to improve our understanding of metformin’s transgenerational effects to develop prophylactic strategies for the prevention of adverse fetal outcomes.
Collapse
|
31
|
A Whole-Body Physiologically Based Pharmacokinetic Model Characterizing Interplay of OCTs and MATEs in Intestine, Liver and Kidney to Predict Drug-Drug Interactions of Metformin with Perpetrators. Pharmaceutics 2021; 13:pharmaceutics13050698. [PMID: 34064886 PMCID: PMC8151202 DOI: 10.3390/pharmaceutics13050698] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 04/30/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Transmembrane transport of metformin is highly controlled by transporters including organic cation transporters (OCTs), plasma membrane monoamine transporter (PMAT), and multidrug/toxin extrusions (MATEs). Hepatic OCT1, intestinal OCT3, renal OCT2 on tubule basolateral membrane, and MATE1/2-K on tubule apical membrane coordinately work to control metformin disposition. Drug–drug interactions (DDIs) of metformin occur when co-administrated with perpetrators via inhibiting OCTs or MATEs. We aimed to develop a whole-body physiologically based pharmacokinetic (PBPK) model characterizing interplay of OCTs and MATEs in the intestine, liver, and kidney to predict metformin DDIs with cimetidine, pyrimethamine, trimethoprim, ondansetron, rabeprazole, and verapamil. Simulations showed that co-administration of perpetrators increased plasma exposures to metformin, which were consistent with clinic observations. Sensitivity analysis demonstrated that contributions of the tested factors to metformin DDI with cimetidine are gastrointestinal transit rate > inhibition of renal OCT2 ≈ inhibition of renal MATEs > inhibition of intestinal OCT3 > intestinal pH > inhibition of hepatic OCT1. Individual contributions of transporters to metformin disposition are renal OCT2 ≈ renal MATEs > intestinal OCT3 > hepatic OCT1 > intestinal PMAT. In conclusion, DDIs of metformin with perpetrators are attributed to integrated effects of inhibitions of these transporters.
Collapse
|
32
|
Ruan Y, Li X, You L, Chen J, Shen Y, Zhang J, Yuan Y, Kang L, Qin C, Wu C. Effect of Pharmaceutical Excipients on Intestinal Absorption of Metformin via Organic Cation-Selective Transporters. Mol Pharm 2021; 18:2198-2207. [PMID: 33956455 DOI: 10.1021/acs.molpharmaceut.0c01104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Growing evidence has shown that some pharmaceutical excipients can act on drug transporters. The present study was aimed at investigating the effects of 13 commonly used excipients on the intestinal absorption of metformin (MTF) and the underlying mechanisms using Caco-2 cells and an ex vivo mouse non-everted gut sac model. First, the uptake of MTF in Caco-2 cells was markedly inhibited by nonionic excipients including Solutol HS 15, polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, and crospovidone. Second, transport profile studies showed that MTF was taken up via multiple cation-selective transporters, among which a novel pyrilamine-sensitive proton-coupled organic cation (H+/OC+) antiporter played a key role. Third, Solutol HS 15, polysorbate 40, and polysorbate 60 showed cis-inhibitory effects on the uptake of either pyrilamine (prototypical substrate of the pyrilamine-sensitive H+/OC+ antiporter) or 1-methyl-4-phenylpyridinium (substrate of traditional cation-selective transporters including OCTs, MATEs, PMAT, SERT, and THTR-2), indicating that their suppression on MTF uptake is due to the synergistic inhibition toward multiple influx transporters. Finally, the pH-dependent mouse intestinal absorption of MTF was significantly decreased by Solutol HS 15, polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, and pyrilamine. In conclusion, this study revealed that a novel transport process mediated by the pyrilamine-sensitive H+/OC+ antiporter contributes to the intestinal absorption of MTF in conjunction with the traditional cation-selective transporters. Mechanistic understanding of the interaction of excipients with cation-selective transporters can improve the formulation design and clinical application of cationic drugs.
Collapse
Affiliation(s)
- Yiling Ruan
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xinran Li
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Linjun You
- Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, Nanjing 211198, China
| | - Jungen Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yueyue Shen
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Junying Zhang
- Department of TCMs Pharmaceuticals, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yaozuo Yuan
- Jiangsu Institute for Food and Drug Control, Nanjing 210019, China
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Chao Qin
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
33
|
Zhou S, Zeng S, Shu Y. Drug-Drug Interactions at Organic Cation Transporter 1. Front Pharmacol 2021; 12:628705. [PMID: 33679412 PMCID: PMC7925875 DOI: 10.3389/fphar.2021.628705] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
The interaction between drugs and various transporters is one of the decisive factors that affect the pharmacokinetics and pharmacodynamics of drugs. The organic cation transporter 1 (OCT1) is a member of the Solute Carrier 22A (SLC22A) family that plays a vital role in the membrane transport of organic cations including endogenous substances and xenobiotics. This article mainly discusses the drug-drug interactions (DDIs) mediated by OCT1 and their clinical significance.
Collapse
Affiliation(s)
- Shiwei Zhou
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, MD, United States.,Department of Thyroid Surgery, The Second Xiangya Hospital, Central South University, Hunan, China
| | - Sujuan Zeng
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China
| | - Yan Shu
- Key Laboratory of Oral Medicine, School and Hospital of Stomatology, Guangzhou Medical University, Guangzhou, China.,Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, Baltimore, MD, United States
| |
Collapse
|
34
|
Yang M, Darwish T, Larraufie P, Rimmington D, Cimino I, Goldspink DA, Jenkins B, Koulman A, Brighton CA, Ma M, Lam BYH, Coll AP, O'Rahilly S, Reimann F, Gribble FM. Inhibition of mitochondrial function by metformin increases glucose uptake, glycolysis and GDF-15 release from intestinal cells. Sci Rep 2021; 11:2529. [PMID: 33510216 PMCID: PMC7843649 DOI: 10.1038/s41598-021-81349-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023] Open
Abstract
Even though metformin is widely used to treat type2 diabetes, reducing glycaemia and body weight, the mechanisms of action are still elusive. Recent studies have identified the gastrointestinal tract as an important site of action. Here we used intestinal organoids to explore the effects of metformin on intestinal cell physiology. Bulk RNA-sequencing analysis identified changes in hexose metabolism pathways, particularly glycolytic genes. Metformin increased expression of Slc2a1 (GLUT1), decreased expression of Slc2a2 (GLUT2) and Slc5a1 (SGLT1) whilst increasing GLUT-dependent glucose uptake and glycolytic rate as observed by live cell imaging of genetically encoded metabolite sensors and measurement of oxygen consumption and extracellular acidification rates. Metformin caused mitochondrial dysfunction and metformin's effects on 2D-cultures were phenocopied by treatment with rotenone and antimycin-A, including upregulation of GDF15 expression, previously linked to metformin dependent weight loss. Gene expression changes elicited by metformin were replicated in 3D apical-out organoids and distal small intestines of metformin treated mice. We conclude that metformin affects glucose uptake, glycolysis and GDF-15 secretion, likely downstream of the observed mitochondrial dysfunction. This may explain the effects of metformin on intestinal glucose utilisation and food balance.
Collapse
Affiliation(s)
- Ming Yang
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Tamana Darwish
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Pierre Larraufie
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Debra Rimmington
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Irene Cimino
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Deborah A Goldspink
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Benjamin Jenkins
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Albert Koulman
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Cheryl A Brighton
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Marcella Ma
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Anthony P Coll
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Stephen O'Rahilly
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Frank Reimann
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Fiona M Gribble
- MRC Metabolic Diseases Unit, Addenbrooke's Hospital, Wellcome Trust/MRC Institute of Metabolic Science (IMS), University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
35
|
Metry M, Shu Y, Abrahamsson B, Cristofoletti R, Dressman JB, Groot DW, Parr A, Langguth P, Shah VP, Tajiri T, Mehta MU, Polli JE. Biowaiver Monographs for Immediate Release Solid Oral Dosage Forms: Metformin Hydrochloride. J Pharm Sci 2021; 110:1513-1526. [PMID: 33450218 DOI: 10.1016/j.xphs.2021.01.011] [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] [Received: 11/10/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 01/11/2023]
Abstract
Data are examined regarding possible waiver of in vivo bioequivalence testing (i.e. biowaiver) for approval of metformin hydrochloride (metformin) immediate-release solid oral dosage forms. Data include metformin's Biopharmaceutics Classification System (BCS) properties, including potential excipient interactions. Metformin is a prototypical transporter-mediated drug and is highly soluble, but only 50% of an orally administered dose is absorbed from the gut. Therefore, metformin is a BCS Class III substance. A BCS-based approval approach for major changes to marketed products and new generics is admissible if test and reference dosage forms have the identical active pharmaceutical ingredient and if in vitro dissolution from both are very rapid (i.e. at least 85% within 15 min at pH 1.2, 4.5, and 6.8). Recent International Council for Harmonisation BCS guidance indicates all excipients for Class III biowaivers are recommended to be qualitatively the same and quantitatively similar (except for preservatives, flavor agents, colorant, or capsule shell or film coating excipients). However, despite metformin being a prototypical transporter-mediated drug, there is no evidence that commonly used excipients impact metformin absorption, such that this restriction on excipients for BCS III drugs merits regulatory relief. Commonly used excipients in usual amounts are not likely to impact metformin absorption.
Collapse
Affiliation(s)
- Melissa Metry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations AstraZeneca, Gothenburg, Sweden
| | - Rodrigo Cristofoletti
- Brazilian Health Surveillance Agency (Anvisa), Division of Bioequivalence, Brasilia, Brazil
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - D W Groot
- RIVM-National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Alan Parr
- Bioceutics LCC, Raleigh-Durham, North Carolina, USA
| | - Peter Langguth
- Department of Pharmaceutical Technology and Biopharmaceutics, Johannes Gutenberg University, Mainz, Germany
| | - Vinod P Shah
- International Pharmaceutical Federation (FIP), The Hague, the Netherlands
| | - Tomokazu Tajiri
- Astellas Pharma Inc, Analytical Research Laboratories, Yaizu, Japan
| | - Mehul U Mehta
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
36
|
Ott M, Werneke U. Wernicke's encephalopathy - from basic science to clinical practice. Part 1: Understanding the role of thiamine. Ther Adv Psychopharmacol 2020; 10:2045125320978106. [PMID: 33447357 PMCID: PMC7780320 DOI: 10.1177/2045125320978106] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/10/2020] [Indexed: 01/19/2023] Open
Abstract
Wernicke's encephalopathy (WE) is an acute neuropsychiatric state. Untreated, WE can lead to coma or death, or progress to Korsakoff syndrome (KS) - a dementia characterized by irreversible loss of anterograde memory. Thiamine (vitamin B1) deficiency lies at the heart of this condition. Yet, our understanding of thiamine regarding prophylaxis and treatment of WE remains limited. This may contribute to the current undertreatment of WE in clinical practice. The overall aim of this review is to identify the best strategies for prophylaxis and treatment of WE in regard to (a) dose of thiamine, (b) mode of administration, (c) timing of switch from one mode of administration to another, (d) duration of administration, and (e) use of magnesium along thiamine as an essential cofactor. Evidence from randomized controlled trials and other intervention studies is virtually absent. Therefore, we have to resort to basic science for proof of principle instead. Here, we present the first part of our clinical review, in which we explore the physiology of thiamine and the pathophysiology of thiamine deficiency. We first explore both of these in their historical context. We then review the pharmacodynamics and pharmacokinetics of thiamine, exploring the roles of the six currently known thiamine compounds, their transporters, and target enzymes. We also explore the significance of magnesium as a cofactor in thiamine-facilitated enzymatic reactions and thiamine transport. In the second (forthcoming) part of this review, we will use the findings of the current review to make evidence-based inferences about strategies for prophylaxis and treatment of WE.
Collapse
Affiliation(s)
- Michael Ott
- Department of Public Health and Clinical Medicine, Division of Medicine, Umeå University, Umeå, Sweden
| | - Ursula Werneke
- Department of Clinical Sciences, Division of Psychiatry, Sunderby Research Unit, Umeå University, Umeå, Sweden
| |
Collapse
|
37
|
Kurosawa K, Chiba K, Noguchi S, Nishimura T, Tomi M. Development of a Pharmacokinetic Model of Transplacental Transfer of Metformin to Predict In Vivo Fetal Exposure. Drug Metab Dispos 2020; 48:1293-1302. [PMID: 33051249 DOI: 10.1124/dmd.120.000127] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022] Open
Abstract
Two types of systems are used in ex vivo human placental perfusion studies to predict fetal drug exposures, that is, closed systems with recirculation of the maternal and fetal buffer and open systems using a single-pass mode without recirculation. The in vivo fetal/maternal (F:M) ratio of metformin, a cationic drug that crosses the placenta, is consistent with that reported in an open system ex vivo but not with that in a closed system. In the present study, we aimed to develop a pharmacokinetic (PK) model of transplacental transfer of metformin to predict in vivo fetal exposure to metformin and to resolve the apparent inconsistency between open and closed ex vivo systems. The developed model shows that the difference between open and closed systems is due to the difference in the time required to achieve the steady state. The model-predicted F:M ratio (approx. 0.88) is consistent with reported in vivo values [mean (95% confidence interval): 1.10 (0.69-1.51)]. The model incorporates bidirectional transport via organic cation transporter 3 (OCT3) at the basal plasma membrane, and simulations indicate that the use of trimethoprim (an OCT3 inhibitor) to prevent microbial growth in the placenta ex vivo has a negligible effect on the overall maternal-to-fetal and fetal-to-maternal clearances. The model could successfully predict in vivo fetal exposure using ex vivo human placental perfusion data from both closed and open systems. This transplacental PK modeling approach is expected to be useful for evaluating human fetal exposures to other poorly permeable compounds, besides metformin. SIGNIFICANCE STATEMENT: We developed a pharmacokinetic model of transplacental transfer of metformin, used to treat gestational diabetes mellitus, in order to predict in vivo fetal exposure and resolve the discrepancy between reported findings in open and closed ex vivo perfusion systems. The discrepancy is due to a difference in the time required to reach the steady state. The model can predict in vivo fetal exposure using data from both closed and open systems.
Collapse
Affiliation(s)
- Ken Kurosawa
- Department of Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Koji Chiba
- Department of Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Saki Noguchi
- Department of Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Tomohiro Nishimura
- Department of Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| | - Masatoshi Tomi
- Department of Clinical Pharmacology, Janssen Pharmaceutical K.K., Tokyo, Japan (K.K.); Laboratory of Clinical Pharmacology, Yokohama University of Pharmacy, Kanagawa, Japan (K.C.); and Faculty of Pharmacy, Keio University of Pharmacy, Tokyo, Japan (K.K., S.N., T.N., M.T.)
| |
Collapse
|
38
|
Nguyen JT, Tian DD, Tanna RS, Hadi DL, Bansal S, Calamia JC, Arian CM, Shireman LM, Molnár B, Horváth M, Kellogg JJ, Layton ME, White JR, Cech NB, Boyce RD, Unadkat JD, Thummel KE, Paine MF. Assessing Transporter-Mediated Natural Product-Drug Interactions Via In vitro-In Vivo Extrapolation: Clinical Evaluation With a Probe Cocktail. Clin Pharmacol Ther 2020; 109:1342-1352. [PMID: 33174626 DOI: 10.1002/cpt.2107] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022]
Abstract
The botanical natural product goldenseal can precipitate clinical drug interactions by inhibiting cytochrome P450 (CYP) 3A and CYP2D6. Besides P-glycoprotein, effects of goldenseal on other clinically relevant transporters remain unknown. Established transporter-expressing cell systems were used to determine the inhibitory effects of a goldenseal extract, standardized to the major alkaloid berberine, on transporter activity. Using recommended basic models, the extract was predicted to inhibit the efflux transporter BCRP and uptake transporters OATP1B1/3. Using a cocktail approach, effects of the goldenseal product on BCRP, OATP1B1/3, OATs, OCTs, MATEs, and CYP3A were next evaluated in 16 healthy volunteers. As expected, goldenseal increased the area under the plasma concentration-time curve (AUC0-inf ) of midazolam (CYP3A; positive control), with a geometric mean ratio (GMR) (90% confidence interval (CI)) of 1.43 (1.35-1.53). However, goldenseal had no effects on the pharmacokinetics of rosuvastatin (BCRP and OATP1B1/3) and furosemide (OAT1/3); decreased metformin (OCT1/2, MATE1/2-K) AUC0-inf (GMR, 0.77 (0.71-0.83)); and had no effect on metformin half-life and renal clearance. Results indicated that goldenseal altered intestinal permeability, transport, and/or other processes involved in metformin absorption, which may have unfavorable effects on glucose control. Inconsistencies between model predictions and pharmacokinetic outcomes prompt further refinement of current basic models to include differential transporter expression in relevant organs and intestinal degradation/metabolism of the precipitant(s). Such refinement should improve in vitro-in vivo prediction accuracy, contributing to a standard approach for studying transporter-mediated natural product-drug interactions.
Collapse
Affiliation(s)
- James T Nguyen
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Dan-Dan Tian
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Rakshit S Tanna
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Deena L Hadi
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA.,Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA
| | - Sumit Bansal
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Justina C Calamia
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Christopher M Arian
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Laura M Shireman
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Bálint Molnár
- SOLVO Biotechnology, SZTE Biológiai Epület, University of Szeged, Szeged, Hungary
| | - Miklós Horváth
- SOLVO Biotechnology, SZTE Biológiai Epület, University of Szeged, Szeged, Hungary
| | - Joshua J Kellogg
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - Matthew E Layton
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, USA
| | - John R White
- Department of Pharmacotherapy, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Nadja B Cech
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA.,Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina, USA
| | - Richard D Boyce
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jashvant D Unadkat
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA.,Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Kenneth E Thummel
- Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA.,Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Mary F Paine
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA.,Center of Excellence for Natural Product Drug Interaction Research, Spokane, Washington, USA
| |
Collapse
|
39
|
Chen X, Wang DD, Li ZP. Time course and dose effect of metformin on weight in patients with different disease states. Expert Rev Clin Pharmacol 2020; 13:1169-1177. [PMID: 32940086 DOI: 10.1080/17512433.2020.1822164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVES The present study was to quantitate and compare the efficacy of metformin on weight in different disease states using model-based meta-analysis (MBMA). METHODS Randomized controlled trials (RCT) of metformin effects on weight in different disease states were collected by searching the public databases. The change rate of weight from baseline was selected as the efficacy indicator. RESULTS A total 21 RCTs containing 1885 patients including patients with type 2 diabetes mellitus, patients with antipsychotic induced weight gain, patients with obesity, were included into the present study. After deducting placebo effect, the maximal effect (Emax) of metformin on weight in patients with type 2 diabetes mellitus, patients with antipsychotic induced weight gain, patients with obesity were -6.86%, -8.82%, and -4.14%, respectively. The treatment duration to reach half of the maximal effect (ET50) were 107, 45.5, and 15.1 weeks, respectively. Within the metformin dose range from 21 RCTs, no significant dose-response relationship was observed. However, the time-course relationship is obvious for efficacy of metformin on weight. CONCLUSIONS The present study firstly provided quantitative information for metformin effects on weight in different disease states, including patients with type 2 diabetes mellitus, patients with antipsychotic induced weight gain, patients with obesity.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Pharmacy, Children's Hospital of Fudan University , Shanghai, China
| | - Dong-Dong Wang
- Department of Pharmacy, Children's Hospital of Fudan University , Shanghai, China
| | - Zhi-Ping Li
- Department of Pharmacy, Children's Hospital of Fudan University , Shanghai, China
| |
Collapse
|
40
|
Li AP. In Vitro Human Cell-Based Experimental Models for the Evaluation of Enteric Metabolism and Drug Interaction Potential of Drugs and Natural Products. Drug Metab Dispos 2020; 48:980-992. [PMID: 32636209 DOI: 10.1124/dmd.120.000053] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/18/2020] [Indexed: 02/13/2025] Open
Abstract
Elements of key enteric drug metabolism and disposition pathways are reviewed to aid the assessment of the applicability of current cell-based enteric experimental systems for the evaluation of enteric metabolism and drug interaction potential. Enteric nuclear receptors include vitamin D receptor, constitutive androstane receptor, pregnane X receptor, farnesoid X receptor, liver X receptor, aryl hydrocarbon receptor, and peroxisome proliferator-activated receptor. Enteric drug metabolizing enzyme pathways include both cytochrome P450 (P450) and non-P450 drug metabolizing enzymes based on gene expression, proteomics, and activity. Both uptake and efflux transporters are present in the small intestine, with P-glycoprotein found to be responsible for most drug-drug and food-drug interactions. The cell-based in vitro enteric systems reviewed are 1) immortalized cell line model: the human colon adenocarcinoma (Caco-2) cells; 2) human stem cell-derived enterocyte models: stem cell enteric systems, either from intestinal crypt cells or induced pluripotent stem cells; and 3) primary cell models: human intestinal slices, cryopreserved human enterocytes, permeabilized cofactor-supplemented (MetMax) cryopreserved human enterocytes, and cryopreserved human intestinal mucosa. The major deficiency with both immortalized cell lines and stem cell-derived enterocytes is that drug metabolizing enzyme activities, although they are detectable, are substantially lower than those for the intestinal mucosa in vivo. Human intestine slices, cryopreserved human enterocytes, MetMax cryopreserved human enterocytes, and cryopreserved human intestinal mucosa retain robust enteric drug metabolizing enzyme activity and represent appropriate models for the evaluation of metabolism and metabolism-dependent drug interaction potential of orally administered xenobiotics including drugs, botanical products, and dietary supplements. SIGNIFICANCE STATEMENT: Enteric drug metabolism plays an important role in the bioavailability and metabolic fate of orally administered drugs as well as in enteric drug-drug and food-drug interactions. The current status of key enteric drug metabolism and disposition pathways and in vitro human cell-based enteric experimental systems for the evaluation of the metabolism and drug interaction potential of orally administered substances is reviewed.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories, Inc., Columbia, Maryland
| |
Collapse
|
41
|
García MA, Contreras D, González PM. Metformin Transport in Native MDCK-Wt and MDCK-II Monolayers Unveils Functional Inter-Strains Differences Influencing Drug Permeability. Pharm Res 2020; 37:121. [PMID: 32514792 DOI: 10.1007/s11095-020-02824-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/14/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE MDCK cells are commonly used to assess drug permeability, but the existence of various strains merits a comparative functional study. Since metformin absorption is largely mediated by transporters and paracellular diffusion, we used it to functionally compare MDCK-wt and MDCK-II. METHODS Uptake, bidirectional transport and efflux experiments were performed using different buffers, pH, and a panel of transporter inhibitors. Relative contributions to total transport in both strains were estimated. RESULTS Metformin uptake into MDCK-wt was linear but saturable in MDCK-II. Uptake into MDCK-wt or -II was promoted at pH 5.4 or 8.4, respectively. Quinidine and cimetidine similarly inhibited uptake in both strains. Lopinavir (PMAT specific) at pH 5.4 or pyrimethamine (MATE specific) at pH 8.4 differentially inhibited MDCK-wt or -II, respectively. Transport at pH 7.4 was absorptive regardless of strains, but secretory (MDCK-II) or absorptive (MDCK-wt) at pH 5.4. Efflux was largely basolateral in both strains. While paracellular permeability was similar between strains, total transport was dominated by transporters in MDCK-II or paracellular diffusion in MDCK-wt. CONCLUSIONS Metformin transport revealed functional differences between MDCK strains. Apical uptake was governed by MATE in MDCK-II or PMAT in MDCK-wt, such that metformin transport was either secretory or absorptive, respectively.
Collapse
Affiliation(s)
- Mauricio A García
- Department of Biopharmaceutics and Pharmaceutical Technology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.,Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Danae Contreras
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile.,Innovation and Biopharmaceutical Evaluation Center (IBECenter), Av. Mexico, #715, Recoleta, Santiago, Chile
| | - Pablo M González
- Innovation and Biopharmaceutical Evaluation Center (IBECenter), Av. Mexico, #715, Recoleta, Santiago, Chile.
| |
Collapse
|
42
|
Liao MZ, Flood Nichols SK, Ahmed M, Clark S, Hankins GD, Caritis S, Venkataramanan R, Haas D, Quinney SK, Haneline LS, Tita AT, Manuck T, Wang J, Thummel KE, Brown LM, Ren Z, Easterling TR, Hebert MF. Effects of Pregnancy on the Pharmacokinetics of Metformin. Drug Metab Dispos 2020; 48:264-271. [PMID: 31980499 PMCID: PMC7076518 DOI: 10.1124/dmd.119.088435] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/30/2019] [Indexed: 12/22/2022] Open
Abstract
This study's primary objective was to fully characterize the pharmacokinetics of metformin in pregnant women with gestational diabetes mellitus (GDM) versus nonpregnant controls. Steady-state oral metformin pharmacokinetics in pregnant women with GDM receiving either metformin monotherapy (n = 24) or a combination with glyburide (n = 30) as well as in nonpregnant women with type 2 diabetes mellitus (T2DM) (n = 24) were determined utilizing noncompartmental techniques. Maternal and umbilical cord blood samples were collected at delivery from 38 women. With both 500- and 1000-mg doses, metformin bioavailability, volume of distribution beta (V β ), clearance, and renal clearance were significantly increased during pregnancy. In addition, in the women receiving metformin 500 mg, significantly higher metformin apparent oral clearance (CL/F) (27%), weight-adjusted renal secretion clearance (64%), and apparent oral volume of distribution beta (V β /F) (33%) were seen during pregnancy. Creatinine clearance was significantly higher during pregnancy. Increasing metformin dose from 500 to 1000 mg orally twice daily significantly increased V β /F by 28%, weight-adjusted V β /F by 32% and CL/F by 25%, and weight-adjusted CL/F by 28% during pregnancy. Mean metformin umbilical cord arterial-to-venous plasma concentration ratio was 1.0 ± 0.1, venous umbilical cord-to-maternal concentration ratio was 1.4 ± 0.5, and arterial umbilical cord-to-maternal concentration ratio was 1.5 ± 0.5. Systemic exposure after a 500-mg dose of metformin was lower during pregnancy compared with the nonpregnant women with T2DM. However, in patients receiving metformin 1000 mg, changes in estimated bioavailability during pregnancy offset the changes in clearance leading to no significant change in CL/F with the higher dose. SIGNIFICANCE STATEMENT: Gestational diabetes mellitus complicates 5%-13% of pregnancies and is often treated with metformin. Pregnant women undergo physiological changes that alter drug disposition. Preliminary data suggest that pregnancy lowers metformin concentrations, potentially affecting efficacy and safety. This study definitively describes pregnancy's effects on metformin pharmacokinetics and expands the mechanistic understanding of pharmacokinetic changes across the dosage range. Here we report the nonlinearity of metformin pharmacokinetics and the increase in bioavailability, clearance, renal clearance, and volume of distribution during pregnancy.
Collapse
Affiliation(s)
- Michael Z Liao
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Shannon K Flood Nichols
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Mahmoud Ahmed
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Shannon Clark
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Gary D Hankins
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Steve Caritis
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Raman Venkataramanan
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - David Haas
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Sara K Quinney
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Laura S Haneline
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Alan T Tita
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Tracy Manuck
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Joanne Wang
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Kenneth E Thummel
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Linda Morris Brown
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Zhaoxia Ren
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Thomas R Easterling
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| | - Mary F Hebert
- University of Washington, Departments of Pharmaceutics (M.Z.L., J.W., K.E.T.), Obstetrics and Gynecology (T.R.E., M.F.H.), and Pharmacy (T.R.E., M.F.H.), Seattle, Washington; Madigan Army Medical Center, Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tacoma, Washington (S.K.F.N.); University of Texas Medical Branch in Galveston, Department of Obstetrics and Gynecology, Galveston, Texas (M.A., S.Cl., G.D.H.); University of Pittsburgh, Departments of Obstetrics and Gynecology (S.Ca.), Pharmacy and Pharmaceutical Sciences (R.V.), Pittsburgh, Pennsylvania; Indiana University, Departments of Obstetrics and Gynecology (D.H., S.K.Q.) and Pediatrics (L.S.H.), Indianapolis, Indiana; University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Birmingham, Alabama (A.T.T.); University of North Carolina, Department of Obstetrics and Gynecology, Chapel Hill, North Carolina (T.M.); Biostatistics and Epidemiology Division, Environmental and Health Science Unit, RTI International, Rockville, Maryland (L.M.B.); and Obstetric and Pediatric Pharmacology and Therapeutic Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (Z.R.)
| |
Collapse
|
43
|
Nikolaidis S, Virgiliou C, Vekiou M, Skari A, Kechagidou A, Gika H, Theodoridis G, Pappas P, Leondaritis G, Mougios V. Effect of exercise on key pharmacokinetic parameters related to metformin absorption in healthy humans: A pilot study. Scand J Med Sci Sports 2020; 30:858-864. [PMID: 31975547 DOI: 10.1111/sms.13628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 01/11/2020] [Accepted: 01/16/2020] [Indexed: 12/14/2022]
Abstract
Exercise is widely accepted as having therapeutic effects; thus, it is important to know whether it interacts with medications. The aim of the present pilot study was to examine the effect of high-intensity interval exercise (known to have antidiabetic action) on key pharmacokinetic parameters related to absorption of metformin (the first-line medication against type 2 diabetes). Ten healthy men participated in two sessions, spaced one to two weeks apart in random, counterbalanced order. In both sessions, participants received 1000 mg of metformin orally, 1-1.5 hours after breakfast. Then, they either ran for 60 minutes at alternating intensity, starting at 40 minutes after metformin administration, and rested without food consumption over the next 3 hours or they rested without food consumption during the entire testing period. Venous blood samples were collected before and at 0.5, 2, 2.5, 3, 3.5, 4, and 4.5 hours after metformin administration for metformin determination by liquid chromatography-mass spectrometry. Capillary blood samples were also collected for lactate and glucose measurements. Data from the two sessions were compared through Wilcoxon or Student's t test, as appropriate. Maximum plasma concentration of metformin (Cmax ) was higher at exercise compared to rest (P = .059). Time to reach Cmax (Tmax ) decreased with exercise (P = .009), and the area under the metformin concentration vs time curve was higher at exercise (P = .047). The addition of exercise to metformin administration did not cause hypoglycemia or lactic acidosis. In conclusion, our results provide the first evidence that pharmacokinetic values related to metformin absorption are affected by exercise.
Collapse
Affiliation(s)
- Stefanos Nikolaidis
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sport Science, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Christina Virgiliou
- BIOMIC_AUTh, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,FoodOmicsGR RI, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,School of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Magdalini Vekiou
- Department of Critical Care and Emergency, General Hospital of Grevena, Grevena, Greece
| | - Ariadni Skari
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sport Science, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Helen Gika
- BIOMIC_AUTh, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,FoodOmicsGR RI, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Theodoridis
- BIOMIC_AUTh, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,FoodOmicsGR RI, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,School of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Periklis Pappas
- Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - George Leondaritis
- Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Vassilis Mougios
- Laboratory of Evaluation of Human Biological Performance, School of Physical Education and Sport Science, Aristotle University of Thessaloniki, Thessaloniki, Greece.,BIOMIC_AUTh, Center for Interdisciplinary Research and Innovation, Thermi, Greece.,FoodOmicsGR RI, Center for Interdisciplinary Research and Innovation, Thermi, Greece
| |
Collapse
|
44
|
Jinakote M, Ontawong A, Soodvilai S, Pimta J, Pasachan T, Chatsudthipong V, Srimaroeng C. High affinity of 4-(4-(dimethylamino)styryl)-N-methylpyridinium transport for assessing organic cation drugs in hepatocellular carcinoma cells. Fundam Clin Pharmacol 2020; 34:365-379. [PMID: 31883148 DOI: 10.1111/fcp.12531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 12/12/2022]
Abstract
Human organic cation transporter 1 (hOCT1) and human organic cation transporter 3 (hOCT3) are highly expressed in hepatocytes and play important roles in cationic drug absorption, distribution, and elimination. A previous study demonstrated that downregulation of hOCT1 and hOCT3 mRNA was related to hepatocellular carcinoma (HepG2) prognosis and severity. Whether these transporters expressed in HepG2 cells serve for cationic drug delivery has not been investigated. Besides radioactive transport, options for assessing hOCTs in hepatocytes are limited. This study clarified the significant roles of hOCTs in HepG2 by comparing cationic fluorescent 4-(4-(dimethylamino)styryl)-N-methylpyridinium (ASP+ ) with traditional [3 H]-1-methyl-4-phenylpyridinium (MPP+ ). The results showed ASP+ was preferably transported into HepG2 compared to [3 H]-MPP+ with high affinity and a high maximal transport rate. Selective transport of ASP+ mediated by hOCTs was influenced by extracellular pH, temperature, and membrane depolarization, corresponding to hOCT1 and hOCT3 expressions. Furthermore, transport of cationic drugs, metformin, and paclitaxel in HepG2 cells was blunted by OCT inhibitors, suggesting that hOCT1 and hOCT3 expressed in HepG2 cells exhibit notable impacts on cationic drug actions. The fluorescent ASP+ -based in vitro model may also provide a rapid and powerful analytical tool for further screening of cationic drug actions and interactions with hOCTs, particularly hOCT1 and hOCT3 in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Metee Jinakote
- Faculty of Oriental Medicine, Chiang Rai College, Chiang Rai, 57000, Thailand.,Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Atcharaporn Ontawong
- Unit of Excellence of Coffee, Division of Physiology, School of Medical Sciences, University of Phayao, Phayao, 56000, Thailand
| | - Sunhapas Soodvilai
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Jeerawat Pimta
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Tipthida Pasachan
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Varanuj Chatsudthipong
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Chutima Srimaroeng
- Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
45
|
Abstract
The organic cation transporters (OCTs) OCT1, OCT2, OCT3, novel OCT (OCTN)1, OCTN2, multidrug and toxin exclusion (MATE)1, and MATE kidney-specific 2 are polyspecific transporters exhibiting broadly overlapping substrate selectivities. They transport organic cations, zwitterions, and some uncharged compounds and operate as facilitated diffusion systems and/or antiporters. OCTs are critically involved in intestinal absorption, hepatic uptake, and renal excretion of hydrophilic drugs. They modulate the distribution of endogenous compounds such as thiamine, L-carnitine, and neurotransmitters. Sites of expression and functions of OCTs have important impact on energy metabolism, pharmacokinetics, and toxicity of drugs, and on drug-drug interactions. In this work, an overview about the human OCTs is presented. Functional properties of human OCTs, including identified substrates and inhibitors of the individual transporters, are described. Sites of expression are compiled, and data on regulation of OCTs are presented. In addition, genetic variations of OCTs are listed, and data on their impact on transport, drug treatment, and diseases are reported. Moreover, recent data are summarized that indicate complex drug-drug interaction at OCTs, such as allosteric high-affinity inhibition of transport and substrate dependence of inhibitor efficacies. A hypothesis about the molecular mechanism of polyspecific substrate recognition by OCTs is presented that is based on functional studies and mutagenesis experiments in OCT1 and OCT2. This hypothesis provides a framework to imagine how observed complex drug-drug interactions at OCTs arise. Finally, preclinical in vitro tests that are performed by pharmaceutical companies to identify interaction of novel drugs with OCTs are discussed. Optimized experimental procedures are proposed that allow a gapless detection of inhibitory and transported drugs.
Collapse
Affiliation(s)
- Hermann Koepsell
- Institute of Anatomy and Cell Biology and Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| |
Collapse
|
46
|
Mutual role of ecto-5'-nucleotidase/CD73 and concentrative nucleoside transporter 3 in the intestinal uptake of dAMP. PLoS One 2019; 14:e0223892. [PMID: 31634358 PMCID: PMC6802847 DOI: 10.1371/journal.pone.0223892] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 09/14/2019] [Indexed: 12/18/2022] Open
Abstract
2'-Deoxyadenosine 5'-monophosphate (dAMP), a deoxyribonucleotide found in DNA, affects intestinal cell growth. The molecular mechanisms underlying gastrointestinal absorption of foreign DNA ingested along with food has hardly been investigated. The aim of this study was to investigate the mechanism underlying intestinal absorption of dAMP. The uptake of [3H]dAMP by Caco-2 cells was Na+- and pH-dependent and was inhibited by various nucleosides. In contrast, nitrobenzylthioinosine (NMBPR), an equilibrative nucleoside transporter inhibitor, showed little inhibitory effects on [3H]dAMP uptake. Additionally, human concentrative nucleoside transporter (CNT) 3, transiently expressed in COS-7 cells, mediated the uptake of [3H]dAMP. A kinetic study revealed that the Km value of CNT3-mediated uptake of dAMP (59.6 μM) was close to that of 2'-deoxyadenosine (dAdo) (56.3 μM), whereas the dAMP Vmax (15.6 pmol·mg protein–1min–1) was 500-fold lesser than the dAdo Vmax (7782 pmol·mg protein–1min–1). Further, [3H]dAMP uptake was greater in COS-7 cells expressing ecto-5'-nucleotidase/CD73 with CNT3 than in those expressing CNT3 alone. These data suggest that, although dAMP is a substrate of CNT3, it is dephosphorylated to dAdo by CD73 and is efficiently absorbed as dAdo from the intestinal lumen.
Collapse
|
47
|
Chen Y, Xue F, Xia G, Zhao Z, Chen C, Li Y, Zhang Y. Transepithelial transport mechanisms of 7,8-dihydroxyflavone, a small molecular TrkB receptor agonist, in human intestinal Caco-2 cells. Food Funct 2019; 10:5215-5227. [PMID: 31384856 DOI: 10.1039/c9fo01007f] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
7,8-Dihydroxyflavone (7,8-DHF), as a high-affinity TrkB receptor agonist, has been extensively explored in many human disorders involving brain-derived neurotrophic factor (BDNF) such as Alzheimer's disease, Parkinson's disease, depression, and obesity. However, to date, the transepithelial transport mechanisms of 7,8-DHF in the intestines remain unclear. The aim of our work was to quantify and to characterize in vitro transport of naturally occurring 7,8-DHF distinguished by its physicochemical and pharmacological properties. We discussed the transport mechanisms of 7,8-DHF using the Caco-2 cell model to determine the bi-directional permeability with different environmental factors (time, concentration, pH, metabolic inhibitors etc.). The influx and efflux characteristics of 7,8-DHF were also clarified. 7,8-DHF was poorly transported across Caco-2 cell monolayers by mainly passive diffusion via a transcellular pathway and not a paracellular pathway. The transport of 7,8-DHF was time and concentration-dependent in both the apical (AP) to basolateral (BL) side and the reverse direction. Interestingly, decreasing the pH from 7.4 to 6.0 markedly enhanced 7,8-DHF transport. It is noteworthy that 7,8-DHF transport was strongly inhibited by metabolic inhibitors and was highly dependent on temperature. The efflux ratio (ER) values at different concentrations were all above 1.5, indicating the existence of the efflux transporter. We found that breast cancer resistance protein (BCRP) was not involved in 7,8-DHF secretion and that the transport mechanism of 7,8-DHF was passive transport with an active efflux mediated by P-glycoprotein (P-gp) and multidrug resistance associated proteins (MRPs), particularly MRP 2. Moreover, the use of various influx transporter inhibitors in Caco-2 cells showed that organic cation transporters (OCTs) and organic anion-transporting polypeptides (OATPs) participated in 7,8-DHF transport. Taken together, the elucidated transport characteristics of 7,8-DHF provide useful information for designing novel and efficient delivery systems and avoiding food-food or food-drug interactions.
Collapse
Affiliation(s)
- Yufeng Chen
- Department of Food Science and Nutrition, School of Biosystems Engineering and Food Science, Zhejiang Key Laboratory for Agro-Food Processing; Zhejiang Engineering Center for Food Technology and Equipment; Zhejiang University, Hangzhou 310058, China.
| | | | | | | | | | | | | |
Collapse
|
48
|
Cai H, Everett RS, Thakker DR. Efficacious dose of metformin for breast cancer therapy is determined by cation transporter expression in tumours. Br J Pharmacol 2019; 176:2724-2735. [PMID: 31032880 DOI: 10.1111/bph.14694] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 04/04/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE It has been extensively reported that the leading anti-diabetic drug, metformin, exerts significant anticancer effects. This hydrophilic, cationic drug requires cation transporters for cellular entry where it activates its intracellular target, the AMPK signalling pathway. However, clinical results on metformin therapy (used at antidiabetic doses) for breast cancer are ambiguous. It is likely that the antidiabetic dose is inadequate in patients that have breast tumours with low cation transporter expression, resulting in non-responsiveness to the drug. We postulate that cation transporter expression and metformin dose are key determinants in its antitumour efficacy in breast cancer. EXPERIMENTAL APPROACH Antitumour efficacy of metformin was compared between low cation transporter-expressing MCF-7 breast tumours and MCF-7 tumours overexpressing organic cation transporter 3 (OCT3-MCF7). A dose-response relationship of metformin in combination with standard-of-care paclitaxel (for oestrogen receptor-positive MCF-7 breast tumours) or carboplatin (for triple-negative MDA-MB-468 breast tumours) was investigated in xenograft mice. KEY RESULTS Metformin had greater efficacy against tumours with higher cation transporter expression, as observed in OCT3-MCF7 versus MCF-7 tumours and MDA-MB-468 versus MCF-7 tumours. In MCF-7 tumours, a threefold higher metformin dose was required to achieve intratumoural exposure that was comparable to exposure in MDA-MB-468 tumours and enhance antitumour efficacy of standard-of-care in MCF-7 tumours versus MDA-MB-468 tumours. Antitumour efficacy correlated with intratumoural AMPK activation and metformin concentration. CONCLUSIONS AND IMPLICATIONS An efficacious metformin dose for breast cancer varies among tumour subtypes based on cation transporter expression, which provides a useful guide for dose selection.
Collapse
Affiliation(s)
- Hao Cai
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ruth S Everett
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dhiren R Thakker
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
49
|
Dawed AY, Zhou K, van Leeuwen N, Mahajan A, Robertson N, Koivula R, Elders PJM, Rauh SP, Jones AG, Holl RW, Stingl JC, Franks PW, McCarthy MI, 't Hart LM, Pearson ER. Variation in the Plasma Membrane Monoamine Transporter (PMAT) (Encoded by SLC29A4) and Organic Cation Transporter 1 (OCT1) (Encoded by SLC22A1) and Gastrointestinal Intolerance to Metformin in Type 2 Diabetes: An IMI DIRECT Study. Diabetes Care 2019; 42:1027-1033. [PMID: 30885951 DOI: 10.2337/dc18-2182] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/11/2019] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Gastrointestinal adverse effects occur in 20-30% of patients with metformin-treated type 2 diabetes, leading to premature discontinuation in 5-10% of the cases. Gastrointestinal intolerance may reflect localized high concentrations of metformin in the gut. We hypothesized that reduced transport of metformin via the plasma membrane monoamine transporter (PMAT) and organic cation transporter 1 (OCT1) could increase the risk of severe gastrointestinal adverse effects. RESEARCH DESIGN AND METHODS The study included 286 severe metformin-intolerant and 1,128 metformin-tolerant individuals from the IMI DIRECT (Innovative Medicines Initiative: DIabetes REsearCh on patient straTification) consortium. We assessed the association of patient characteristics, concomitant medication, and the burden of mutations in the SLC29A4 and SLC22A1 genes on odds of intolerance. RESULTS Women (P < 0.001) and older people (P < 0.001) were more likely to develop metformin intolerance. Concomitant use of transporter-inhibiting drugs increased the odds of intolerance (odds ratio [OR] 1.72, P < 0.001). In an adjusted logistic regression model, the G allele at rs3889348 (SLC29A4) was associated with gastrointestinal intolerance (OR 1.34, P = 0.005). rs3889348 is the top cis-expression quantitative trait locus for SLC29A4 in gut tissue where carriers of the G allele had reduced expression. Homozygous carriers of the G allele treated with transporter-inhibiting drugs had more than three times higher odds of intolerance compared with carriers of no G allele and not treated with inhibiting drugs (OR 3.23, P < 0.001). Use of a genetic risk score derived from rs3889348 and SLC22A1 variants found that the odds of intolerance were more than twice as high in individuals who carry three or more risk alleles compared with those carrying none (OR 2.15, P = 0.01). CONCLUSIONS These results suggest that intestinal metformin transporters and concomitant medications play an important role in the gastrointestinal adverse effects of metformin.
Collapse
Affiliation(s)
- Adem Y Dawed
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, U.K
| | - Kaixin Zhou
- Division of Population Health and Genomics, School of Medicine, University of Dundee, Dundee, U.K
| | - Nienke van Leeuwen
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anubha Mahajan
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Neil Robertson
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K
| | - Robert Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K.,Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Skåne University Hospital, Malmö, Lund University, Malmö, Sweden
| | - Petra J M Elders
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Simone P Rauh
- Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Angus G Jones
- Institute of Clinical and Biological Sciences, University of Exeter Medical School, Exeter, U.K
| | - Reinhard W Holl
- Institute of Epidemiology and Medical Biometry (ZIBMT), University of Ulm, Ulm, Germany, and German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Julia C Stingl
- Research Division, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Skåne University Hospital, Malmö, Lund University, Malmö, Sweden.,Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Mark I McCarthy
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K.,Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, U.K.,Oxford National Institute for Health Research Biomedical Research Centre, Oxford University Hospitals Trust, Oxford, U.K
| | - Leen M 't Hart
- Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam Public Health Research Institute, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Section of Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
50
|
Ningrum VDA, Istikharah R, Firmansyah R. Allele Frequency of SLC22A1 Met420del Metformin Main Transporter Encoding Gene among Javanese-Indonesian Population. Open Access Maced J Med Sci 2019; 7:378-383. [PMID: 30834005 PMCID: PMC6390162 DOI: 10.3889/oamjms.2019.087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Genetic variation in the genes that encode metformin transporters has been proven to cause pharmacokinetic variability and various glycemic response to metformin. Organic Cation Transporter (OCT) 1 protein encoded by the SLC22A1 gene is primarily responsible for the process of metformin influx to the hepatocytes as the target of antihyperglycemic action as well as metformin elimination through the renal. This study aimed to determine the allele frequency distribution of the SLC22A1 Met420del gene in OCT1 among the Javanese population, the largest ethnic group in Indonesia with T2DM. METHODS The research involved 100 adult patients from 9 healthcare facilities in Yogyakarta Province. The PCR-RFLP method was employed as a genotype analysis to detect polymorphism using 5'-AGGTTCACGGACTCTGTGCT-3' forward primer and 5'-AAGCTGGAGTGTGCGATCT-3' reverse primer. RESULTS No AA variant (wild type) type was found in the SLC22A1 Met420del gene, and only 4% of the subjects had Aa heterozygote type. The allele frequencies of A and a were 2.0% and 98.0% in all subjects, respectively. CONCLUSION The allele frequencies in the Javanese-Indonesian population were almost the same as those in the studies involving Japanese, Chinese-Han, and Asian-American populations. This study recommends further research on the correlation between the influence of methionine deletion at codon 420 on the variability of pharmacokinetic profiles and the glycemic response to metformin as well as the incidence of gastrointestinal intolerance due to metformin administration.
Collapse
Affiliation(s)
- Vitarani DA Ningrum
- Laboratory of Pharmaceutical Research, Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta, Indonesia
| | - Rochmy Istikharah
- Laboratory of Biochemistry, Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta, Indonesia
| | - Rheza Firmansyah
- Department of Pharmacy, Universitas Islam Indonesia, Yogyakarta, Indonesia
| |
Collapse
|