Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Hear Res ; 447: 109013, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718672

ABSTRACT

Cisplatin, a highly effective chemotherapeutic drug for various human cancers, induces irreversible sensorineural hearing loss as a side effect. Currently there are no highly effective clinical strategies for the prevention of cisplatin-induced ototoxicity. Previous studies have indicated that short-term cisplatin ototoxicity primarily affects the outer hair cells of the cochlea. Therefore, preventing the entry of cisplatin into hair cells may be a promising strategy to prevent cisplatin ototoxicity. This study aimed to investigate the entry route of cisplatin into mouse cochlear hair cells. The competitive inhibitor of organic cation transporter 2 (OCT2), cimetidine, and the sensory mechanoelectrical transduction (MET) channel blocker benzamil, demonstrated a protective effect against cisplatin toxicity in hair cells in cochlear explants. Sensory MET-deficient hair cells explanted from Tmc1Δ;Tmc2Δ mice were resistant to cisplatin toxicity. Cimetidine showed an additive protective effect against cisplatin toxicity in sensory MET-deficient hair cells. However, in the apical turn, cimetidine, benzamil, or genetic ablation of sensory MET channels showed limited protective effects, implying the presence of other entry routes for cisplatin to enter the hair cells in the apical turn. Systemic administration of cimetidine failed to protect cochlear hair cells from ototoxicity caused by systemically administered cisplatin. Notably, outer hair cells in MET-deficient mice exhibited no apparent deterioration after systemic administration of cisplatin, whereas the outer hair cells in wild-type mice showed remarkable deterioration. The susceptibility of mouse cochlear hair cells to cisplatin ototoxicity largely depends on the sensory MET channel both ex vivo and in vivo. This result justifies the development of new pharmaceuticals, such as a specific antagonists for sensory MET channels or custom-designed cisplatin analogs which are impermeable to sensory MET channels.


Subject(s)
Antineoplastic Agents , Cimetidine , Cisplatin , Mechanotransduction, Cellular , Organic Cation Transporter 2 , Ototoxicity , Cisplatin/toxicity , Animals , Ototoxicity/prevention & control , Ototoxicity/metabolism , Ototoxicity/physiopathology , Mechanotransduction, Cellular/drug effects , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/antagonists & inhibitors , Cimetidine/pharmacology , Antineoplastic Agents/toxicity , Hair Cells, Auditory/drug effects , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Hair Cells, Auditory, Outer/drug effects , Hair Cells, Auditory, Outer/pathology , Hair Cells, Auditory, Outer/metabolism , Mice, Inbred C57BL , Mice , Membrane Proteins
2.
Arch Toxicol ; 98(7): 2131-2142, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38589558

ABSTRACT

Oxaliplatin (OHP) is effective in colorectal cancer treatment but induces peripheral neurotoxicity (OHP-induced peripheral neurotoxicity, OIPN), diminishing survivor quality of life. Organic cation transporter 2 (OCT2) is a key OHP uptake pathway in dorsal root ganglia. Competing for OCT2-mediated OHP uptake, such as with the tyrosine kinase inhibitor dasatinib, may mitigate OHP side effects. We investigated OHP and dasatinib interaction with OCT2 in human embryonic kidney 293 (HEK293) cells expressing OCT2 within a 10-3 to 10-7 M concentration range. Uptake competition experiments using fluorescent organic cation 4-(4-dimethylaminostyryl)-N-methylpyridinium (ASP+, 1 µM) and mass spectrometry (MS) to determine cellular platinum content indicated that OHP (100 µM) is an OCT2 substrate, mediating OHP cellular toxicity. ASP+ and MS analysis revealed dasatinib as a non-transported inhibitor of hOCT2 (IC50 = 5.9 µM) and as a regulator of OCT2 activity. Dasatinib reduced transporter Vmax, potentially via Y544 phosphorylation suppression. MS analysis showed cellular dasatinib accumulation independent of hOCT2. Although 3 µM dasatinib reduced 100 µM OHP accumulation in hOCT2-HEK293 cells, co-incubation with dasatinib and OHP did not prevent OHP toxicity, possibly due to dasatinib-induced cell viability reduction. In summary, this study demonstrates OHP as an OCT2 substrate and dasatinib as a non-transported inhibitor and regulator of OCT2, offering potential for OIPN mitigation.


Subject(s)
Antineoplastic Agents , Dasatinib , Organic Cation Transporter 2 , Oxaliplatin , Protein Kinase Inhibitors , Humans , Dasatinib/pharmacology , HEK293 Cells , Oxaliplatin/pharmacology , Organic Cation Transporter 2/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Antineoplastic Agents/toxicity , Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/toxicity , Drug Interactions , Pyridinium Compounds/pharmacology
3.
J Pharm Pharmacol ; 74(8): 1180-1192, 2022 Aug 19.
Article in English | MEDLINE | ID: mdl-35704284

ABSTRACT

OBJECTIVES: To explore clonidine (Clon) nephroprotective effects as an inhibitor of organic cationic transporter 2 (OCT2) and p38 mitogen-activated protein kinase (p38 MAPK) against cisplatin (CP)-induced nephrotoxicity. OCT2 is mainly responsible for renal accumulation of CP. Clon has been recently recognized as an OCT2 inhibitor and exerts beneficial effects on renal function and p38 MAPK. This study further investigates its underlying anti-inflammatory, antioxidative and antiapoptotic effects. METHODS: Rats were randomly assigned into five groups: (I) CON, (II) CP, (III) CP + Clon 0.125, (IV) CP + Clon 0.25, (V) CP + Clon 0.5, and (VI) Clon 0.5 alone. Clon was administered orally at 0.125, 0.25 and 0.5 mg/kg/day dosages for 10 days. On day 7, rats in groups from (II) to (V) received a single intraperitoneal injection of CP (10 mg/kg). KEY FINDINGS: Clon 0.25 mg/kg displayed the best nephroprotective outcomes, justified by the significant amelioration of parameters like renal function, oxidative stress, and inflammatory status, as well as modulated the OCT2 expression, phosphorylation of p38 and p53, compared with Clon 0.125 and 0.5 mg/kg. CONCLUSION: This study suggests the promising nephroprotective impact of Clon as an OCT2 inhibitor against CP nephrotoxicity and its proficient role in attenuating oxidative stress, inflammatory status and apoptotic status.


Subject(s)
Antineoplastic Agents , Cisplatin , Clonidine , Organic Cation Transporter 2 , p38 Mitogen-Activated Protein Kinases , Animals , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Clonidine/pharmacology , Kidney/drug effects , Organic Cation Transporter 2/antagonists & inhibitors , Oxidative Stress , Rats , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
4.
Int J Mol Sci ; 23(3)2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35163125

ABSTRACT

The organic cation transporters OCT1-3 (SLC22A1-3) facilitate the transport of cationic endo- and xenobiotics and are important mediators of drug distribution and elimination. Their polyspecific nature makes OCTs highly susceptible to drug-drug interactions (DDIs). Currently, screening of OCT inhibitors depends on uptake assays that require labeled substrates to detect transport activity. However, these uptake assays have several limitations. Hence, there is a need to develop novel assays to study OCT activity in a physiological relevant environment without the need to label the substrate. Here, a label-free impedance-based transport assay is established that detects OCT-mediated transport activity and inhibition utilizing the neurotoxin MPP+. Uptake of MPP+ by OCTs induced concentration-dependent changes in cellular impedance that were inhibited by decynium-22, corticosterone, and Tyrosine Kinase inhibitors. OCT-mediated MPP+ transport activity and inhibition were quantified on both OCT1-3 overexpressing cells and HeLa cells endogenously expressing OCT3. Moreover, the method presented here is a valuable tool to identify novel inhibitors and potential DDI partners for MPP+ transporting solute carrier proteins (SLCs) in general.


Subject(s)
Electric Impedance , Gene Expression Regulation/drug effects , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2/metabolism , 1-Methyl-4-phenylpyridinium/adverse effects , Biological Transport , Biological Transport, Active , HEK293 Cells , Herbicides/adverse effects , Humans , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 1/antagonists & inhibitors , Organic Cation Transporter 1/genetics , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/genetics
5.
Hum Exp Toxicol ; 40(12_suppl): S447-S459, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34592875

ABSTRACT

The balance of cisplatin uptake and efflux, mediated mainly by organic cation transporter 2 (OCT2) and multidrug and toxin extrusion 1 (MATE1), respectively, determines the renal accumulation and nephrotoxicity of cisplatin. Using transporter-mediated cellular uptake assay, we identified wedelolactone (WEL), a medicinal plant-derived natural compound, is a competitive inhibitor of OCT2 and a noncompetitive inhibitor of MATE1. Wedelolactone showed a selectivity to inhibit OCT2 rather than MATE1. Cytotoxicity studies revealed that wedelolactone alleviated cisplatin-induced cytotoxicity in OCT2-overexpressing HEK293 cells, whereas it did not alter the cytotoxicity of cisplatin in various cancer cell lines. Additionally, wedelolactone altered cisplatin pharmacokinetics, reduced kidney accumulation of cisplatin, and ameliorated cisplatin-induced acute kidney injury in the Institute of Cancer Research mice. In conclusion, these findings suggest a translational potential of WEL as a natural therapy for preventing cisplatin-induced nephrotoxicity and highlight the need for drug-drug interaction investigations of WEL with other treatments which are substrates of OCT2 and/or MATE1.


Subject(s)
Cisplatin/toxicity , Coumarins/pharmacology , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism , Animals , Antineoplastic Agents/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred ICR , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/genetics
6.
Toxicology ; 459: 152853, 2021 07.
Article in English | MEDLINE | ID: mdl-34252480

ABSTRACT

Oxaliplatin (OXA) is a third-generation platinum drug; however, its application is greatly limited due to the severe peripheral neurotoxicity. This study aims to confirm the transport mechanism of OXA and to explore whether L-tetrahydropalmatine (L-THP) would alleviate OXA-induced peripheral neurotoxicity by selectively inhibiting these uptake transporters in vitro and in vivo. Our results revealed that organic cation transporter 2 (OCT2), organic cation/carnitine transporter 1 (OCTN1) and organic cation/carnitine transporter 2 (OCTN2) were involved in the uptake of OXA in dorsal root ganglion (DRG) neurons and mitochondria, respectively. L-THP (1-100 µM) reduced OXA (40 µM) induced cytotoxicity in MDCK-hOCT2 (Madin-Darby canine kidney, MDCK), MDCK-hOCTN1, MDCK-hOCTN2, and rat primary DRG cells, and decreased the accumulation of OXA in above cells and rat DRG mitochondria, but did not affect its efflux from MDCK-hMRP2 cells. Furthermore, Co-administration of L-THP (5-20 mg/kg for mice, 10-40 mg/kg for rats; twice a week, iv or ig) attenuated OXA (8 mg/kg for mice, 4 mg/kg for rats; twice a week, iv) induced peripheral neurotoxicity and reduced the platinum concentration in the DRG. Whereas, L-THP (1-100 µM for cells; 10-20 mg/kg for mice) did not impair the antitumour efficacy of OXA (40 µM for cells; 8 mg/kg for mice) in HT29 tumour-bearing nude mice nor in tumour cells (HT29 and SW620 cells). In conclusion, OCT2, OCTN1 and OCTN2 contribute to OXA uptake in the DRG and mitochondria. L-THP attenuates OXA-induced peripheral neurotoxicity via inhibiting OXA uptake but without impairing the antitumour efficacy of OXA. L-THP is a potential candidate drug to attenuate OXA-induced peripheral neurotoxicity.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Berberine Alkaloids/pharmacology , Carrier Proteins/antagonists & inhibitors , Ganglia, Spinal/metabolism , Mitochondria/metabolism , Oxaliplatin/pharmacokinetics , Oxaliplatin/toxicity , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control , Animals , Behavior, Animal/drug effects , Dogs , Ganglia, Spinal/drug effects , HEK293 Cells , HT29 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred ICR , Mice, Nude , Mitochondria/drug effects , Multidrug Resistance-Associated Protein 2 , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Rats , Solute Carrier Family 22 Member 5/antagonists & inhibitors , Solute Carrier Family 22 Member 5/metabolism , Symporters/antagonists & inhibitors , Symporters/metabolism
7.
Mol Pharmacol ; 100(4): 348-355, 2021 10.
Article in English | MEDLINE | ID: mdl-34330821

ABSTRACT

Cisplatin is a platinum-based drug, which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing antitumor efficacy. SIGNIFICANCE STATEMENT: Cisplatin-induced hearing loss is a dose-limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Drug Delivery Systems/trends , Hearing Loss/chemically induced , Hearing Loss/metabolism , Membrane Transport Proteins/metabolism , Hearing Loss/prevention & control , Humans , Monocarboxylic Acid Transporters/antagonists & inhibitors , Monocarboxylic Acid Transporters/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism
8.
Pharmacology ; 106(7-8): 390-399, 2021.
Article in English | MEDLINE | ID: mdl-33979803

ABSTRACT

INTRODUCTION: Alzheimer's disease (AD) is a severe neurodegenerative disorder of the brain characterized by degeneration of cholinergic neurons which is directly linked to cognitive decline. Nerve growth factor (NGF) is the most potent protective factor for cholinergic neurons, additionally the NMDA antagonist memantine blocks glutamate-mediated excitotoxic activity. Quinidine is an inhibitor of organic cation transporter 2 (OCT2). OCT2 is located on cholinergic neurons and plays a role in presynaptic reuptake and recycling of acetylcholine in the brain. We hypothesize that quinidine can modulate the protective effects of NGF and memantine on cholinergic neurons in organotypic brain slices of the nucleus basalis of Meynert (nBM). METHODS: Organotypic brain slices of nBM were incubated with 100 ng/mL NGF, 10 µM memantine, 10 µM quinidine, and combinations of these treatments for 2 weeks. Cholinergic neurons were immunohistochemically stained for choline acetyltransferase (ChAT). RESULTS: Our data show that NGF as well as memantine counteracted the cell death of cholinergic nBM neurons. Quinidine alone had no toxic effect on cholinergic neurons but inhibited the protective effect of NGF and memantine when applied simultaneously. DISCUSSION/CONCLUSION: Our data provide evidence that quinidine modulates the survival of cholinergic nBM neurons via OCT2.


Subject(s)
Memantine/pharmacology , Nerve Growth Factor/pharmacology , Neuroprotective Agents/pharmacology , Quinidine/pharmacology , Acetylcholine/metabolism , Animals , Basal Nucleus of Meynert/drug effects , Basal Nucleus of Meynert/metabolism , Brain/drug effects , Brain/metabolism , Cell Survival/drug effects , Cholinergic Neurons , Mice , Mice, Inbred C57BL , Organic Cation Transporter 2/antagonists & inhibitors , Tissue Culture Techniques
9.
J Clin Pharmacol ; 61(4): 461-471, 2021 04.
Article in English | MEDLINE | ID: mdl-32989831

ABSTRACT

Tucatinib is a potent tyrosine kinase inhibitor selective for human epidermal growth factor receptor 2 (HER2) approved by the US Food and Drug Administration for the treatment of HER2-positive metastatic breast cancer and in development for other HER2-positive solid tumors. Modest, reversible serum creatinine (SCr) elevations have been observed in tucatinib clinical trials. SCr is conveyed by the renal drug transporters organic cation transporter 2 (OCT2) and multidrug and toxin extrusion protein 1 (MATE1) and 2-K (MATE2-K) and can increase in the presence of inhibitors of these transporters. In vitro, tucatinib inhibited OCT2-, MATE1-, and MATE2-K-mediated transport of metformin, with IC50 values of 14.7, 0.340, and 0.135 µM, respectively. Tucatinib also inhibited OCT2- and MATE1-mediated transport of creatinine, with IC50 values of 0.107 and 0.0855 µM, respectively. A phase 1 study with metformin administered orally in the absence and presence of tucatinib was conducted in 18 healthy subjects. Renal function was assessed by measuring glomerular filtration rate (GFR; based on iohexol plasma clearance) and endogenous markers (SCr, cystatin C-based estimated glomerular filtration rate [eGFR]) with and without tucatinib. Metformin exposure increased (1.4-fold) and renal clearance decreased (29.99-17.64 L/h) with tucatinib, with no effect on metformin maximum concentration. Creatinine clearance transiently decreased 23% with tucatinib. GFR and eGFR, which are unaffected by OCT2 and/or MATE1/2-K transport, were unchanged with tucatinib. These data demonstrate that tucatinib inhibits OCT2- and MATE1/2-K-mediated tubular secretion of creatinine, which may manifest as mild SCr elevations that are not indicative of renal impairment.


Subject(s)
Antineoplastic Agents/pharmacology , Metformin/pharmacokinetics , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transporter 2/antagonists & inhibitors , Oxazoles/pharmacology , Pyridines/pharmacology , Quinazolines/pharmacology , Adolescent , Adult , Aged , Animals , Biological Transport/drug effects , Creatinine/blood , Cross-Over Studies , Dogs , Female , Glomerular Filtration Rate , HEK293 Cells , Healthy Volunteers , Humans , Inhibitory Concentration 50 , Madin Darby Canine Kidney Cells , Male , Middle Aged , Receptor, ErbB-2/antagonists & inhibitors , Young Adult
11.
Biol Pharm Bull ; 43(11): 1693-1698, 2020.
Article in English | MEDLINE | ID: mdl-33132314

ABSTRACT

Cisplatin is a widely used chemotherapy for solid tumors; however, its benefits are limited by serious nephrotoxicity, particularly in proximal tubular cells. The present study investigated the renoprotective effect and mechanisms of germacrone, a bioactive terpenoid compound found in Curcuma species on cisplatin-induced toxicity of renal cells. Germacrone (50 and 100 µM) attenuated apoptosis of human renal proximal tubular cells, RPTEC/TERT1 following treatment with 50 µM cisplatin and for 48 h. Co-treating RPTEC/TERT1 cells with cisplatin and germacrone significantly reduced cellular platinum content compared with cisplatin treatment alone. The effect of germacrone on organic cation transporter 2 (OCT2) which is a transporter responsible for cisplatin uptake was determined. Germacrone showed an inhibitory effect on OCT2-mediated methyl-4-phenylpyridinium acetate (3H-MPP+) uptake with IC50 of 15 µM with less effect on OCT1. The germacrone's protective effect on cisplatin-induced cytotoxicity was not observed in cancer cells; cisplatin's anti-cancer activity was preserved. In conclusion, germacrone prevents cisplatin-induced toxicity in renal proximal tubular cells via inhibition OCT2 transport function and reducing cisplatin accumulation. Thus germacrone may be a good candidate agent used for reducing cisplatin-induced nephrotoxicity.


Subject(s)
Acute Kidney Injury/prevention & control , Cisplatin/adverse effects , Kidney Tubules, Proximal/drug effects , Organic Cation Transporter 2/antagonists & inhibitors , Sesquiterpenes, Germacrane/pharmacology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/pathology , Octamer Transcription Factor-1/metabolism , Organic Cation Transporter 2/metabolism , Sesquiterpenes, Germacrane/therapeutic use
12.
Drug Metab Dispos ; 48(12): 1303-1311, 2020 12.
Article in English | MEDLINE | ID: mdl-33020068

ABSTRACT

Drug-induced kidney injury is a major clinical problem and causes drug attrition in the pharmaceutical industry. To better predict drug-induced kidney injury, kidney in vitro cultures with enhanced physiologic relevance are developed. To mimic the proximal tubule, the main site of adverse drug reactions in the kidney, human-derived renal proximal tubule epithelial cells (HRPTECs) were injected in one of the channels of dual-channel Nortis chips and perfused for 7 days. Tubes of HRPTECs demonstrated expression of tight junction protein 1 (zona occludens-1), lotus lectin, and primary cilia with localization at the apical membrane, indicating an intact proximal tubule brush border. Gene expression of cisplatin efflux transporters multidrug and toxin extrusion transporter (MATE) 1 (SLC47A1) and MATE2-k (SLC47A2) and megalin endocytosis receptor increased 19.9 ± 5.0-, 23.2 ± 8.4-, and 106 ± 33-fold, respectively, in chip cultures compared with 2-dimensional cultures. Moreover, organic cation transporter 2 (OCT2) (SLC22A2) was localized exclusively on the basolateral membrane. When infused from the basolateral compartment, cisplatin (25 µM, 72 hours) induced toxicity, which was evident as reduced cell number and reduced barrier integrity compared with vehicle-treated chip cultures. Coexposure with the OCT2 inhibitor cimetidine (1 mM) abolished cisplatin toxicity. In contrast, infusion of cisplatin from the apical compartment did not induce toxicity, which was in line with polarized localization of cisplatin uptake transport proteins, including OCT2. In conclusion, we developed a dual channel human kidney proximal tubule-on-a-chip with a polarized epithelium, restricting cisplatin sensitivity to the basolateral membrane and suggesting improved physiologic relevance over single-compartment models. Its implementation in drug discovery holds promise to improve future in vitro drug-induced kidney injury studies. SIGNIFICANCE STATEMENT: Human-derived kidney proximal tubule cells retained characteristics of epithelial polarization in vitro when cultured in the kidney-on-a-chip, and the dual-channel construction allowed for drug exposure using the physiologically relevant compartment. Therefore, cell polarization-dependent cisplatin toxicity could be replicated for the first time in a kidney proximal tubule-on-a-chip. The use of this physiologically relevant model in drug discovery has potential to aid identification of safe novel drugs and contribute to reducing attrition rates due to drug-induced kidney injury.


Subject(s)
Acute Kidney Injury/chemically induced , Cisplatin/toxicity , Kidney Tubules, Proximal/drug effects , Lab-On-A-Chip Devices , Acute Kidney Injury/pathology , Acute Kidney Injury/prevention & control , Cell Culture Techniques/instrumentation , Cells, Cultured , Cimetidine/pharmacology , Cimetidine/therapeutic use , Cisplatin/pharmacokinetics , Drug Evaluation, Preclinical/instrumentation , Feasibility Studies , Gene Expression Profiling , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism
13.
Eur J Drug Metab Pharmacokinet ; 45(6): 725-733, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32860624

ABSTRACT

BACKGROUND AND OBJECTIVE: Imeglimin is a novel oral antidiabetic drug to treat type 2 diabetes, targeting the mitochondrial bioenergetics. In vitro, imeglimin was shown to be a substrate of human multidrug and toxic extrusion transporters MATE1 and MATE2-K and organic cation transporters OCT1 and OCT2. The objective of the study was to assess the potential drug-drug interaction between imeglimin and cimetidine, a reference inhibitor of these transporters. METHODS: A phase 1 study was carried out in 16 subjects who received a single dose of 1500 mg imeglimin alone on day 1 followed by a 6-day treatment (day 5 to day 10) with cimetidine 400 mg twice daily. On day 8, a single dose of imeglimin was co-administered with cimetidine. Blood and urine samples were collected up to 72 h after each imeglimin administration. Pharmacokinetic parameters were determined using non-compartmental methods. RESULTS: Imeglimin maximum plasma concentration (Cmax) and area under the plasma concentration-time curve (AUC) were 1.3-fold [90% CI (1.12-1.62) and (1.10-1.46) for Cmax and AUC0-last, respectively] higher when imeglimin was co-administered with cimetidine but this increase was not considered clinically relevant. This increase could be mainly explained by a reduction in renal elimination, mediated through the cimetidine inhibition of renal MATE1 transporter. Imeglimin taken alone or with cimetidine was safe and well tolerated in all subjects. CONCLUSIONS: No clinically significant drug-drug interaction exists between imeglimin and cimetidine, a reference inhibitor of MATE1, MATE2-K, OCT1 and OCT2 transporters. CLINICAL TRIAL REGISTRATION: EudraCT 2018-001103-36.


Subject(s)
Cimetidine/pharmacology , Histamine H2 Antagonists/pharmacology , Hypoglycemic Agents/pharmacokinetics , Kidney/metabolism , Triazines/pharmacokinetics , Adult , Area Under Curve , Drug Interactions , Female , HEK293 Cells , Healthy Volunteers , Humans , Kidney/drug effects , Male , Middle Aged , Octamer Transcription Factor-1/antagonists & inhibitors , Octamer Transcription Factor-1/metabolism , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism , Young Adult
14.
Biopharm Drug Dispos ; 41(6): 239-247, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32473602

ABSTRACT

Cisplatin is used widely for the treatment of multiple solid tumors. Cisplatin-induced nephrotoxicity is caused by renal accumulation of cisplatin via human organic cation transporter 2 (hOCT2). As lansoprazole, a proton pump inhibitor, is known to inhibit hOCT2 activity, lansoprazole might ameliorate cisplatin-induced nephrotoxicity. A previous study showed that concomitant lansoprazole administration ameliorated nephrotoxicity in patients receiving cisplatin. However, the detailed mechanism remains to be clarified. In the present study, the drug-drug interaction between lansoprazole and cisplatin was examined using hOCT2-expressing cultured cells and rat renal slices. Moreover, the effect of lansoprazole on cisplatin-induced nephrotoxicity and the pharmacokinetics of cisplatin in rats was investigated. In the uptake study, lansoprazole potently inhibited the uptake of cisplatin in hOCT2-expressing cultured cells and rat renal slices. The in vivo rat study showed that concomitant lansoprazole significantly ameliorated cisplatin-induced nephrotoxicity and reduced the renal accumulation of platinum up to approximately 60% of cisplatin alone at 72 h after cisplatin intraperitoneal administration. Furthermore, the renal uptake of platinum at 3 min after intravenous cisplatin administration in rats with cisplatin and lansoprazole decreased to 78% of rats with cisplatin alone. In addition, there was no significant difference in the plasma platinum concentration between rats treated with and without lansoprazole at 3 min after cisplatin intravenous administration. These findings suggested that concomitant lansoprazole ameliorated cisplatin-induced nephrotoxicity by inhibiting rOCT2-mediated cisplatin uptake in rats, thus decreasing cisplatin accumulation in the kidney. The present findings provided important information for the establishment of novel protective approaches to minimize cisplatin-induced nephrotoxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Kidney Diseases/drug therapy , Lansoprazole/therapeutic use , Organic Cation Transporter 2/antagonists & inhibitors , Protective Agents/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cisplatin/pharmacokinetics , HEK293 Cells , Humans , Kidney/drug effects , Kidney/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Lansoprazole/pharmacology , Male , Organic Cation Transporter 2/metabolism , Protective Agents/pharmacology , Rats, Wistar
15.
Biochem Pharmacol ; 177: 114021, 2020 07.
Article in English | MEDLINE | ID: mdl-32389634

ABSTRACT

Cisplatin is a first-line chemotherapeutic agent that is widely used for treatment of various solid tumors. However, cisplatin-induced adverse effects, particularly severe nephrotoxicity, preclude its application. In this study, we showed that L-tetrahydropalmatine (L-THP) could selectively inhibit organic cation transporter 2 (OCT2), which plays a crucial role in renal cisplatin uptake from the circulation. Additionally, we demonstrated that L-THP attenuated cisplatin-induced toxicity in mouse primary renal tubular cells. Subsequently, we verified that L-THP reduced the renal accumulation of cisplatin and alleviated cisplatin-induced renal injury in healthy and tumor-bearing nude mice. In healthy mice, co-treatment of L-THP at 5-40 mg/kg reduced cisplatin renal accumulation to 75.0%-49.9% of that in cisplatin alone group (10 mg/kg), and alleviated cisplatin-induced nephrotoxicity. Additionally, it did not alter Pt concentration in the tumor tissue and did not impair its antitumor efficacy in tumor bearing nude mice. The tumor inhibitory rates of cisplatin (10 mg/kg) co-treated with L-THP at 10, 20 and 40 mg/kg were 71.4%, 70.4% and 69.4%, respectively, in H460 tumor bearing nude mice, higher than that of in cisplatin alone group (60.6%), while in HCT116 tumor bearing nude mice, the tumor inhibitory rates in co-treated with 20 mg/kg L-THP was 34.7% (vs 26.3% in cisplatin alone group). Moreover, L-THP reduced cisplatin accumulation and alleviated cisplatin-induced cytotoxicity in human primary renal tubular cells. Therefore, our findings suggested that concomitant administration of L-THP could attenuate cisplatin-induced renal injury via selective inhibition of OCT2 without impairing its antitumor efficacy.


Subject(s)
Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Antineoplastic Agents/adverse effects , Berberine Alkaloids/pharmacology , Cisplatin/adverse effects , Organic Cation Transporter 2/antagonists & inhibitors , Acute Kidney Injury/metabolism , Animals , Antineoplastic Agents/pharmacokinetics , Cells, Cultured , Cisplatin/pharmacokinetics , Dogs , Kidney Tubules/cytology , Madin Darby Canine Kidney Cells , Male , Mice, Inbred ICR , Mice, Nude , Organic Cation Transport Proteins/antagonists & inhibitors , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/metabolism , Protective Agents/pharmacology , Xenograft Model Antitumor Assays
16.
CPT Pharmacometrics Syst Pharmacol ; 9(6): 310-321, 2020 06.
Article in English | MEDLINE | ID: mdl-32441889

ABSTRACT

Creatinine is the most common clinical biomarker of renal function. As a substrate for renal transporters, its secretion is susceptible to inhibition by drugs, resulting in transient increase in serum creatinine and false impression of damage to kidney. Novel physiologically based models for creatinine were developed here and (dis)qualified in a stepwise manner until consistency with clinical data. Data from a matrix of studies were integrated, including systems data (common to all models), proteomics-informed in vitro-in vivo extrapolation of all relevant transporter clearances, exogenous administration of creatinine (to estimate endogenous synthesis rate), and inhibition of different renal transporters (11 perpetrator drugs considered for qualification during creatinine model development and verification on independent data sets). The proteomics-informed bottom-up approach resulted in the underprediction of creatinine renal secretion. Subsequently, creatinine-trimethoprim clinical data were used to inform key model parameters in a reverse translation manner, highlighting best practices and challenges for middle-out optimization of mechanistic models.


Subject(s)
Creatinine/blood , Kidney Tubules, Proximal/metabolism , Models, Biological , Organic Cation Transporter 2/metabolism , Pharmaceutical Preparations/blood , Renal Elimination , Biomarkers/blood , Drug Monitoring , Glomerular Filtration Rate , HEK293 Cells , Humans , Kidney Tubules, Proximal/drug effects , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/genetics , Permeability , Pharmacokinetics , Proteome , Renal Elimination/drug effects
17.
Eur J Clin Pharmacol ; 76(3): 431-437, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31836927

ABSTRACT

PURPOSE: Lucerastat is an orally available glucosylceramide synthase inhibitor with a potential to provide substrate reduction therapy for Fabry patients independent of their α-galactosidase A genotype. In humans, lucerastat is mainly eliminated as unchanged parent compound through renal excretion both by active secretion and passive filtration. In vitro studies indicated that lucerastat is a substrate of human organic cation transporter 2 (OCT2) mainly expressed in the kidney. METHODS: Therefore, this clinical study, conducted in 14 healthy male subjects, investigated the effect of 800 mg twice-daily oral administration of the OCT2 inhibitor cimetidine at steady state on the single-dose pharmacokinetics (PK) of 500 mg lucerastat. The safety and tolerability of lucerastat administered alone and concomitantly with cimetidine were also evaluated. RESULTS: Exposure to lucerastat was slightly higher upon co-administration of cimetidine indicated by geometric mean area under the plasma concentration-time curve from zero to infinity (AUC0-∞) ratio of 1.22 (90% confidence interval [CI] 1.16-1.28). Cimetidine delayed the time to reach maximum lucerastat concentrations (tmax) by 1 h but did not affect its elimination half-life (t½) or maximum plasma concentration (Cmax) as geometric mean ratios were 1.00 (0.91-1.10) and 1.04 (0.92-1.17), respectively, at cimetidine steady state. Lucerastat was safe and well tolerated when given alone and in combination with cimetidine. CONCLUSION: These results indicate that the single-dose PK of lucerastat are not changed to a clinically relevant extent by cimetidine-mediated OCT2 inhibition, allowing the concomitant use of OCT2 inhibitors with lucerastat without any need for dose adjustment. TRIAL REGISTRATION: EudraCT: 2017-003725-14; ClinicalTrials.gov: NCT03380455.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Cimetidine/therapeutic use , Glucosyltransferases/antagonists & inhibitors , Organic Cation Transporter 2/antagonists & inhibitors , 1-Deoxynojirimycin/pharmacokinetics , Administration, Oral , Adult , Area Under Curve , Healthy Volunteers , Humans , Kidney/drug effects , Kidney/metabolism , Male , Young Adult
18.
Biochem Pharmacol ; 171: 113731, 2020 01.
Article in English | MEDLINE | ID: mdl-31783011

ABSTRACT

Stereoselectivity is well described for receptor binding and enzyme catalysis, but so far has only been scarcely investigated in carrier-mediated membrane transport. We thus studied transport kinetics of racemic (anti)adrenergic drugs by the organic cation transporters OCT1 (wild-type and allelic variants), OCT2, OCT3, MATE1, and MATE2-K with a focus on stereospecificity. OCT1 showed stereoselective uptake with up to 2-fold higher vmax over their corresponding counterpart enantiomers for (R,R)-fenoterol, (R,R)-formoterol, (S)-salbutamol, (S)-acebutolol, and (S)-atenolol. Orciprenaline and etilefrine were also transported stereoselectively. The Km was 2.1-fold and 1.5-fold lower for the (S,S)-enantiomers of fenoterol and formoterol, while no significant difference in Km was seen for the other aforementioned drugs. Common OCT1 variants showed similar enantiopreference to wild-type OCT1, with a few notable exceptions (e.g. a switch in enantiospecificity for fenoterol in OCT1*2 compared to the wild-type). Other cation transporters showed strong differences to OCT1 in stereoselectivity and transport activity: The closely related OCT2 displayed a 20-fold higher vmax for (S,S)-fenoterol compared to (R,R)-fenoterol and OCT2 and OCT3 showed 3.5-fold and 4.6-fold higher vmax for the pharmacologically active (R)-salbutamol over (S)-salbutamol. MATE1 and MATE2-K generally mediated transport with a higher capacity but lower affinity compared to OCT1, with moderate stereoselectivity. Our kinetic studies showed that significant stereoselectivity exists in solute carrier-mediated membrane transport of racemic beta-adrenergic drugs with surprising, and in some instances even opposing, preferences between closely related organic cation transporters. This may be relevant for drug therapy, given the strong involvement of these transporters in hepatic and renal drug elimination.


Subject(s)
Adrenergic Agonists/pharmacology , Adrenergic Antagonists/pharmacology , Organic Cation Transport Proteins/agonists , Organic Cation Transport Proteins/antagonists & inhibitors , Acebutolol/chemistry , Acebutolol/metabolism , Acebutolol/pharmacology , Adrenergic Agonists/chemistry , Adrenergic Agonists/metabolism , Adrenergic Antagonists/chemistry , Adrenergic Antagonists/metabolism , Atenolol/chemistry , Atenolol/pharmacology , Biological Transport , Fenoterol/chemistry , Fenoterol/metabolism , Fenoterol/pharmacology , Formoterol Fumarate/chemistry , Formoterol Fumarate/metabolism , Formoterol Fumarate/pharmacology , HEK293 Cells , Humans , Kinetics , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 1/agonists , Organic Cation Transporter 1/antagonists & inhibitors , Organic Cation Transporter 1/metabolism , Organic Cation Transporter 2/agonists , Organic Cation Transporter 2/antagonists & inhibitors , Organic Cation Transporter 2/metabolism , Stereoisomerism
19.
Am J Physiol Renal Physiol ; 317(3): F720-F734, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31313952

ABSTRACT

Organic cation transporter 2 (OCT2) clears the blood of cationic drugs. Efforts to understand OCT2 selectivity as a means to predict the potential of new molecular entities (NMEs) to produce unwanted drug-drug interactions typically assess the influence of the NMEs on inhibition of transport. However, the identity of the substrate used to assess transport activity can influence the quantitative profile of inhibition. Metformin and 1-methyl-4-phenylpyridinium (MPP), in particular, display markedly different inhibitory profiles, with IC50 values for inhibition of MPP transport often being more than fivefold greater than IC50 values for the inhibition of metformin transport by the same compound, suggesting that interaction of metformin and MPP with OCT2 cannot be restricted to competition for a single binding site. Here, we determined the kinetic basis for the mutual inhibitory interaction of metformin and MPP with OCT2 expressed in Chinese hamster ovary cells. Although metformin did produce simple competitive inhibition of MPP transport, MPP was a mixed-type inhibitor of metformin transport, decreasing the maximum rate of mediated substrate transport and increasing the apparent Michaelis constant (Ktapp) for OCT2-mediated metformin transport. Furthermore, whereas the IC50 value for metformin's inhibition of MPP transport did not differ from the Ktapp value for metformin transport, the IC50 value for MPP's inhibition of metformin transport was less than its Ktapp value for transport. The simplest model to account for these observations required the influence of a distinct inhibitory site for MPP that, when occupied, decreases the translocation of substrate. These observations underscore the complexity of ligand interaction with OCT2 and argue for use of multiple substrates to obtain the needed kinetic assessment of NME interactions with OCT2.


Subject(s)
1-Methyl-4-phenylpyridinium/pharmacology , Metformin/pharmacology , Organic Cation Transporter 2/antagonists & inhibitors , 1-Methyl-4-phenylpyridinium/metabolism , Animals , Binding Sites , Binding, Competitive , CHO Cells , Cricetulus , Drug Interactions , Kinetics , Metformin/metabolism , Models, Biological , Models, Molecular , Organic Cation Transporter 2/chemistry , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/metabolism , Protein Binding , Protein Conformation
20.
Xenobiotica ; 49(10): 1237-1243, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30472912

ABSTRACT

1. Jatrorrhizine is an active ingredient found in various traditional Chinese medicinal plants. Based on our previous finding that jatrorrhizine was a potent inhibitor of OCT2 and OCT3, the aim of the present study was to explore whether jatrorrhizine has an antidepressant-like action action via inhibition of uptake-2 transporters. 2. In vitro uptake tests showed that jatrorrhizine strongly inhibited PMAT-mediated MPP+ uptake with an IC50 value of 1.05 µM and reduced 5-HT and NE uptake mediated by hOCT2, hOCT3 and hPMAT with IC50 values of 0.1-1 µM (for OCT2 and OCT3) and 1-10 µM (for PMAT). 3. In mouse synaptosomes, jatrorrhizine suppressed 5-HT and NE uptake in a concentration dependently manner, where the role of uptake-2 inhibition is significant. 4. The antidepressant-like action of jatrorrhizine was evaluated by mouse tail suspension test (TST). The TST showed that one week of jatrorrhizine (5, 10 and 20 mg/kg, i.p.) or venlafaxine (20 mg/kg, i.g.) can significantly reduce the duration of immobility when compared with vehicle control group. 5. The concentration of jatrorrhizine shows a dose-dependent increase in brain tissues. 6. Our study suggested that jatrorrhizine might be used as an antidepressant agent via inhibition of uptake-2 transporters.


Subject(s)
Antidepressive Agents , Berberine/analogs & derivatives , Organic Cation Transporter 2/antagonists & inhibitors , Serotonin/metabolism , Animals , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , Berberine/pharmacokinetics , Berberine/pharmacology , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred ICR , Organic Cation Transporter 2/metabolism , Venlafaxine Hydrochloride/pharmacokinetics , Venlafaxine Hydrochloride/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL