Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Pharmaceuticals (Basel) ; 16(9)2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37765032

ABSTRACT

Most of the currently available drugs are derived from natural sources, but they are used only after extensive chemical modifications to improve their safety and efficacy. Natural products are used in health supplements and cosmetic preparations and have been used as auxiliary drugs or alternative medicines. When used in combination with conventional drugs, these herbal products are known to alter their pharmacokinetics and pharmacodynamics, reducing their therapeutic effects. Moreover, herb-drug interactions (HDIs) may have serious side effects, which is one of the major concerns in health practice. It is postulated that HDIs affect the pathways regulating cytochrome P450 enzymes (CYPs). Betanin, the chief pigment of red beetroot (Beta vulgaris L.), has various types of pharmacological activity, such as anti-inflammatory, antioxidant, and anticancer effects. However, the potential risk of HDIs for betanin has not yet been studied. Thus, we aimed to predict more specific HDIs by evaluating the effects of betanin on CYPs (CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, and CYP3A4), the major phase I metabolic enzymes, using fluorescence-/luminescence-based assays. Our results showed that betanin inhibited CYP3A4 activity in a dose-dependent manner (IC50 = 20.97 µΜ). Moreover, betanin acted as a competitive inhibitor of CYP3A4, as confirmed by evaluating Lineweaver-Burk plots (Ki value = 19.48 µΜ). However, no significant inhibitory effects were observed on other CYPs. Furthermore, betanin had no significant effect on CYP1A2, CYP2B6, or CYP2C9 induction in HepG2 cells. In conclusion, betanin acted as a competitive inhibitor of CYP3A4, and thus it should be used cautiously with other drugs that require metabolic enzymes as substrates. Additional in vivo studies and clinical trials are needed to further elucidate the HDIs of betanin.

2.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768840

ABSTRACT

Obesity is a lipid metabolism disorder caused by genetic, medicinal, nutritional, and other environmental factors. It is characterized by a complex condition of excess lipid accumulation in adipocytes. Adipogenesis is a differentiation process that converts preadipocytes into mature adipocytes and contributes to excessive fat deposition. Saikosaponin A (SSA) and saikosaponin D (SSD) are triterpenoid saponins separated from the root of the Bupleurum chinensis, which has long been used to treat inflammation, fever, and liver diseases. However, the effects of these constituents on lipid accumulation and obesity are poorly understood. We investigated the anti-obesity effects of SSA and SSD in mouse 3T3-L1 adipocytes. The MTT assay was performed to measure cell viability, and Oil Red O staining was conducted to determine lipid accumulation. Various adipogenic transcription factors were evaluated at the protein and mRNA levels by Western blot assay and quantitative reverse transcription polymerase chain reaction (qRT-PCR). Here, we showed that SSA and SSD significantly inhibited lipid accumulation without affecting cell viability within the range of the tested concentrations (0.938-15 µM). SSA and SSD also dose-dependently suppressed the expression of peroxisome proliferator-activated receptor gamma (PPARγ), CCAAT/enhancer binding protein alpha (C/EBPα), sterol regulatory element binding protein-1c (SREBP-1c), and adiponectin. Furthermore, the decrease of these transcriptional factors resulted in the repressed expression of several lipogenic genes including fatty acid binding protein (FABP4), fatty acid synthase (FAS), and lipoprotein lipase (LPL). In addition, SSA and SSD enhanced the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) and its substrate, acetyl-CoA carboxylase (ACC), and inhibited the phosphorylation of extracellular-regulated kinase 1/2 (ERK1/2) and p38, but not c-Jun-N-terminal kinase (JNK). These results suggest that SSA and SSD inhibit adipogenesis through the AMPK or mitogen-activated protein kinase (MAPK) pathways in the early stages of adipocyte differentiation. This is the first study on the anti-adipogenic effects of SSA and SSD, and further research in animals and humans is necessary to confirm the potential of saikosaponins as therapeutic agents for obesity.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adenylate Kinase/drug effects , Adenylate Kinase/metabolism , Adipogenesis/genetics , Adiponectin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Obesity Agents/pharmacology , Bupleurum , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Gene Expression/drug effects , Lipogenesis/physiology , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/drug effects , Mitogen-Activated Protein Kinases/metabolism , Obesity/drug therapy , Oleanolic Acid/pharmacology , PPAR gamma/drug effects , PPAR gamma/metabolism , Phosphorylation/drug effects , Plant Extracts/pharmacology , Signal Transduction/physiology , Sterol Regulatory Element Binding Protein 1/metabolism
3.
Endocrinology ; 161(10)2020 10 01.
Article in English | MEDLINE | ID: mdl-32852547

ABSTRACT

Postsurgical hypoparathyroidism is the most common complication of thyroid surgery. Conventional therapy with high-dose calcium and vitamin D can correct hypocalcemia but can increase the risk of hypercalciuria, renal stones, or ectopic calcification. The aim of the present study was to investigate the efficacy of a calcium-sensing receptor antagonist, also called a calcilytic (AXT914), in rat models of postsurgical hypoparathyroidism. Two postsurgical hypoparathyroidism rat models were made by hemi-parathyroidectomy or total parathyroidectomy with autotransplantation in 10-week-old female Wistar rats. AXT914 or vehicle was administered orally for 2 to 3 weeks. Serum PTH, calcium, and phosphorus levels, and the urinary excretion of calcium were measured. Autotransplanted parathyroid tissues were collected and examined histologically. In the hemi-parathyroidectomy model, the oral administration of the calcilytic AXT914 (5 and 10 mg/kg) for 2 weeks increased serum PTH and calcium levels and decreased serum phosphorus levels and urinary calcium excretion. In the total parathyroidectomy with autotransplantation model, the oral administration of AXT914 (10 mg/kg) for 3 weeks increased serum PTH and calcium levels and decreased serum phosphorus levels. The serum PTH and calcium levels increased by AXT914 were maintained for 1 week, even after discontinuation of the drug. In conclusion, AXT914 increased PTH secretion in rat models of postsurgical hypoparathyroidism, thereby correcting abnormal calcium and phosphorus homeostasis. Furthermore, AXT914 improved the functional recovery of autotransplanted parathyroid tissues.


Subject(s)
Hypoparathyroidism/drug therapy , Postoperative Complications/drug therapy , Quinazolinones/administration & dosage , Animals , Combined Modality Therapy , Disease Models, Animal , Drug Administration Schedule , Female , Hypercalciuria/etiology , Hypercalciuria/prevention & control , Hypoparathyroidism/etiology , Hypoparathyroidism/pathology , Parathyroid Glands/transplantation , Parathyroidectomy/adverse effects , Parathyroidectomy/methods , Postoperative Complications/etiology , Postoperative Complications/pathology , Postoperative Period , Rats , Rats, Wistar , Therapies, Investigational , Thyroid Diseases/surgery , Thyroidectomy/adverse effects , Transplantation, Autologous
4.
Curr Drug Deliv ; 16(1): 86-92, 2019.
Article in English | MEDLINE | ID: mdl-30246640

ABSTRACT

BACKGROUND: Chrysin is a strong inhibitor of breast cancer resistance protein (BCRP) but it is practically insoluble in water. Effective solubilization of chrysin is critical for its pharmaceutical application as an absorption enhancer via inhibition of BCRP-mediated drug efflux. OBJECTIVE: This study aimed to develop an effective oral formulation of chrysin to improve its in vivo effect as an absorption enhancer. METHOD: Solid dispersions (SDs) of chrysin were prepared with hydrophilic carriers having surface acting properties and a pH modulator. In vitro and in vivo characterizations were performed to select the optimal SDs of chrysin. RESULTS: SDs with Brij®L4 and aminoclay was most effective in increasing the solubility of chrysin by 13-53 fold at varying drug-carrier ratios. Furthermore, SDs significantly improved the dissolution rate and extent of drug release. SDs (chrysin: Brij®L4: aminoclay=1:3:5) achieved approximately 60% and 83% drug release within 1 h and 8 h, respectively, in aqueous medium, while the dissolution of the untreated chrysin was less than 13%. XRD patterns indicated the amorphous state of chrysin in SDs. The SD formulation was effective in improving the bioavailability of topotecan, a BCRP substrate in rats. Following oral administration of topotecan with the SDs of chrysin, the Cmax and AUC of topotecan was enhanced by approximately 2.6- and 2-fold, respectively, while the untreated chrysin had no effect. CONCLUSION: The SD formulation of chrysin with Brij®L4 and aminoclay appeared to be promising in improving the dissolution of chrysin and enhancing its in vivo effect as an absorption enhancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Flavonoids/pharmacology , Intestinal Absorption/drug effects , Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Administration, Oral , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Availability , Clay/chemistry , Detergents/chemistry , Drug Compounding/methods , Drug Evaluation, Preclinical , Flavonoids/chemistry , Flavonoids/therapeutic use , HT29 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Male , Polidocanol/chemistry , Rats , Rats, Sprague-Dawley , Solubility , Topotecan/pharmacology , Topotecan/therapeutic use , Water/chemistry
5.
J Pharm Biomed Anal ; 115: 272-6, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26263054

ABSTRACT

A rapid, sensitive and selective liquid chromatography-tandem mass spectrometric method (LC-MS/MS) was developed for the quantification of biflorin in rat plasma. Using naringin as an internal standard, plasma samples were subjected to a direct protein precipitation process using methanol. Chromatographic separation was achieved on a Gemini C18 column with an isocratic mobile phase consisting of 0.1% formic acid and methanol (50:50, v/v) at a flow rate of 0.5mL/min. Biflorin was analyzed in the multiple reaction monitoring mode with negative electrospray ionization. The precursor/product ion pairs were m/z 353.0/205.0 and m/z 579.0/271.0 for biflorin and the IS, respectively. The calibration curve was linear over the concentration range of 5-2000ng/mL. The intra- and inter-day precision was less than 7.3% and the accuracy ranged from 96.5 to 103.3%. No significant variation was observed in the stability tests. This method was successfully applied to a pharmacokinetic study of biflorin after the intravenous and oral administration of biflorin to rats. The half-life and oral bioavailability of biflorin were determined as 3.4h and 43%, respectively. This is the first report on the quantitative determination of biflorin in rat plasma as well as the pharmacokinetic characterization of biflorin, which should provide a meaningful foundation for further preclinical and clinical applications of biflorin.


Subject(s)
Chromatography, Liquid/methods , Naphthoquinones/blood , Plant Extracts/pharmacokinetics , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Animals , Injections, Intravenous , Limit of Detection , Male , Naphthoquinones/administration & dosage , Naphthoquinones/isolation & purification , Plant Extracts/administration & dosage , Plant Extracts/isolation & purification , Rats, Sprague-Dawley , Reproducibility of Results
6.
Anticancer Res ; 32(10): 4445-52, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23060571

ABSTRACT

AIM: This study aimed to evaluate the effect of honokiol and its structural analogs on the functional activity and gene expression of P-glycoprotein (P-gp) in order to identify effective P-gp inhibitors from natural products which have additional health-promoting effects. MATERIALS AND METHODS: The interaction characteristics of honokiol, magnolol and 4-O-methylhonokiol with P-gp were determined in NCI/ADR-RES cells overexpressing P-gp. RESULTS: All three compounds down-regulated the expression of P-gp in a concentration- and time-dependent manner, leading to 2.5- to 4.1-fold reductions of P-gp expression in NCI/ADR-RES cells. Accordingly, down-regulation of P-gp resulted in the significant enhancement of the intracellular accumulation of calcein, a P-gp substrate. Furthermore, pre-treatment with honokiol, magnolol or 4-O-methylhonokiol significantly increased the susceptibility of cancer cells to daunorubicin-induced cytotoxicity in NCI/ADR-RES cells. CONCLUSION: The present study suggests that honokiol, magnolol and 4-O-methylhonokiol could be promising agents for reducing the multidrug resistance of cancer cells to anticancer drugs via the down-regulation of P-gp expression.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antineoplastic Agents, Phytogenic/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , Magnolia/chemistry , Plant Extracts/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Daunorubicin/pharmacology , Down-Regulation/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans
7.
Arch Pharm Res ; 34(11): 1865-77, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22139686

ABSTRACT

Over the past few decades, a tremendous amount of work has been done on the molecular characterization of transport proteins in animals and humans, leading to a better understanding of the physiological roles of a number of transport proteins. Furthermore, there is increasing preclinical and clinical evidence to support the importance of transport proteins in the pharmacokinetics and toxicokinetics of a wide variety of structurally diverse drugs. As a consequence, the degree of expression and functionality of transport proteins may directly affect the therapeutic effectiveness, safety and target specificity of drugs. Recently, there has also been increased awareness about potential drug-drug, drug-herb and drug-food interactions involving transporters. Traditionally, a change in metabolic clearance of a drug, particularly via cytochrome P450-mediated metabolism, has been considered the cause of many clinically important drug interactions. However, increasing evidence suggests that some drug interactions result from changes in the activity and/or expression of drug transporters. Accordingly, assessment of the clinical relevance of transporter-mediated drug interactions has become a regulatory issue during the drug approval process and also the evaluation of drug interaction potential has become an integral part of risk assessment during drug development processes. Therefore, this review will highlight the role of some selected drug transporters in drug interactions, as well as their clinical implication.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Drug Interactions , Intestines/drug effects , Kidney/drug effects , Liver/drug effects , Organic Anion Transporters/metabolism , Pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Transport/drug effects , Food-Drug Interactions , Gene Expression Regulation/drug effects , Herb-Drug Interactions , Humans , Intestinal Mucosa/metabolism , Kidney/metabolism , Liver/metabolism , Organic Anion Transporters/antagonists & inhibitors , Organic Anion Transporters/genetics , Plant Preparations/adverse effects , Plant Preparations/pharmacology
8.
Int J Pharm ; 415(1-2): 89-94, 2011 Aug 30.
Article in English | MEDLINE | ID: mdl-21645596

ABSTRACT

The present study aimed to improve the bioavailability of biochanin A, a poorly soluble bioflavonoid, via the preparation of solid dispersion (SD) using Solutol HS15 and HPMC 2910. Solubility of biochanin A was enhanced by 8-60 folds as the drug-carrier ratio was increased in SDs. Furthermore, compared to pure biochanin A or physical mixture (PM), SDs significantly improved the dissolution rate and the extent of drug release. Particularly, SDs (Drug:Solutol HS15:HPMC 2910=1:5:5 or 1:10:10) achieved the rapid and complete drug release (approximately 100% within 1h) at pH 6.8. The XRD patterns indicated that SDs might enhance the solubility of biochanin A by changing the drug crystallinity to amorphous state in addition to the solubilizing effect of hydrophilic carriers. The improved dissolution of biochanin A via SD formulation appeared to be well correlated with the enhanced oral exposure of biochanin A in rats. After an oral administration of SD (Drug:Solutol HS15:HPMC 2910=1:10:10), C(max) and AUC of biochanin A were increased by approximately 13 and 5 folds, respectively, implying that SDs could be effective to improve the bioavailability of biochanin A. In conclusion, solid dispersion with Solutol HS15 and HPMC 2910 appeared to be promising to improve the dissolution and oral exposure of biochanin A.


Subject(s)
Drug Compounding/methods , Genistein , Phytoestrogens , Administration, Oral , Animals , Biological Availability , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Stability , Genistein/administration & dosage , Genistein/chemistry , Genistein/pharmacokinetics , Hydrophobic and Hydrophilic Interactions , Male , Phytoestrogens/administration & dosage , Phytoestrogens/chemistry , Phytoestrogens/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility , X-Ray Diffraction
9.
Mol Pharmacol ; 78(5): 855-64, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713551

ABSTRACT

The transition from a chemotherapy-responsive cancer to a chemotherapy-resistant one is accompanied by increased expression of multidrug resistance 1 (MDR1, p-glycoprotein), which plays an important role in the efflux from the target cell of many anticancer agents. We recently showed that a Forkhead box-containing protein of the O subfamily 1 (FoxO1) is a key regulator of MDR1 gene transcription. Because nuclear localization of FoxO1 is regulated by silent information regulator two ortholog 1 (SIRT1) deacetylase, we wondered whether SIRT1 dominates MDR1 gene expression in breast cancer cells. Overexpression of SIRT1 enhanced both FoxO reporter activity and nuclear levels of FoxO1. Protein expression of MDR1 and gene transcriptional activity were also up-regulated by SIRT1 overexpression. In addition, SIRT1 inhibition reduced both nuclear FoxO1 levels and MDR1 expression in doxorubicin-resistant breast cancer cells (MCF-7/ADR) cells. A potent SIRT1 inhibitor, amurensin G (from Vitis amurensis), was identified by screening plant extracts and bioassay-guided fractionation. The compound suppressed FoxO1 activity and MDR1 expression in MCF-7/ADR cells. Moreover, pretreatment of MCF-7/ADR cells with 1 µg/ml amurensin G for 24 h increased cellular uptake of doxorubicin and restored the responsiveness of MCF-7/ADR cells to doxorubicin. In xenograft studies, injection of 10 mg/kg i.p. amurensin G substantially restored the ability of doxorubicin to inhibit MCF-7/ADR-induced tumor growth. These results suggest that SIRT1 is a potential therapeutic target of MDR1-mediated chemoresistance and that it may be possible to develop amurensin G as a useful agent for chemoresistance reversal.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antibiotics, Antineoplastic/pharmacology , Dibenzocycloheptenes/pharmacology , Doxorubicin/pharmacology , Resorcinols/pharmacology , Sirtuin 1/antagonists & inhibitors , Stilbenes/pharmacology , Animals , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Humans , Methanol , Mice , Mice, Nude , Neoplasm Transplantation , Plant Extracts/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous , Vitis
10.
Eur J Pharm Sci ; 41(2): 226-31, 2010 Oct 09.
Article in English | MEDLINE | ID: mdl-20600879

ABSTRACT

This study investigated the effect of macelignan on the P-glycoprotein-mediated drug efflux as well as CYP3A4-mediated drug metabolism and subsequently its in vivo implication on the bioavailability of paclitaxel. The inhibition effect of macelignan on the CYP3A4-mediated metabolism was negligible over the concentration range of 0.01-100muM in rat liver microsome while approximately 33% inhibition was observed at 100muM in human liver microsome, implying that the interaction of macelignan with CYP3A4 might be insignificant at the physiologically achievable concentrations. In contrast, macelignan (20muM) increased the cellular accumulation of paclitaxel by approximately 1.7-fold in NCI/ADR-RES cells overexpressing P-gp, while it did not alter the cellular accumulation of paclitaxel in OVCAR-8 cells lacking P-gp. The effect of macelignan on the systemic exposure of paclitaxel was also examined in rats after the intravenous and oral administration of paclitaxel in the presence and the absence of macelignan. The concurrent use of macelignan significantly (p<0.05) enhanced the oral exposure of paclitaxel in rats while it did not affect the intravenous pharmacokinetics of paclitaxel, implying that macelignan might be more effective to improve the intestinal absorption rather than reducing hepatic elimination. In conclusion, macelignan appeared to be effective to improve the cellular accumulation as well as oral exposure of paclitaxel mainly via the inhibition of P-gp-mediated cellular efflux, suggesting that the concomitant use of macelignan may provide a therapeutic benefit in improving the anticancer efficacy of paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lignans/pharmacology , Paclitaxel/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Area Under Curve , Blotting, Western , Cytochrome P-450 CYP3A/metabolism , Drug Evaluation, Preclinical , Half-Life , Humans , Lignans/pharmacokinetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Paclitaxel/pharmacokinetics , Rats
11.
Eur J Pharm Sci ; 39(4): 224-32, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20035865

ABSTRACT

Coenzyme Q(10) (CoQ(10)) was formulated into self-nanoemulsifying drug delivery systems (SNEDDS) to overcome low bioavailability attributed to hydrophobic nature of the drug. Screening of oil phase, surfactants and co-surfactants were performed to select Witepsol H35, Solutol HS15 and Lauroglycol FCC, respectively. Ternary phase diagrams were drawn to identify nanoemulsifying region followed by optimization of SNEDDS formulation. The optimized formulation, CoQ(10), Witepsol H35, Solutol HS15 and Lauroglycol FCC in the weight ratio of 1:0.7:4:2, respectively, emulsified readily at 37 degrees C with mean emulsion droplet size of 32.4 nm. The stability test of the optimized formulation in pH 1.2 and 6.8 buffers confirmed no pH effect on emulsion droplet size. In vitro dissolution (emulsification) test and in vivo animal study of the formulation elucidated the complete emulsification of drug and improved oral bioavailability of poorly soluble CoQ(10).


Subject(s)
Drug Delivery Systems/methods , Emulsifying Agents/administration & dosage , Nanotechnology , Triglycerides/administration & dosage , Ubiquinone/analogs & derivatives , Animals , Drug Evaluation, Preclinical/methods , Emulsifying Agents/blood , Emulsifying Agents/chemical synthesis , Male , Nanotechnology/methods , Rats , Rats, Sprague-Dawley , Triglycerides/blood , Triglycerides/chemical synthesis , Ubiquinone/administration & dosage , Ubiquinone/blood , Ubiquinone/chemical synthesis
12.
Nutr Cancer ; 61(4): 538-43, 2009.
Article in English | MEDLINE | ID: mdl-19838926

ABSTRACT

The effect of macelignan, a phytoestrogen, on P-gp function was investigated using multidrug resistant cancer cells overexpressing P-gp (NCI/ADR-RES) and the fluorescent P-gp substrates, daunorubicin and rhodamine 123. Macelignan (40 microM) increased the cellular accumulation of daunorubicin by approximately threefold in NCI/ADR-RES cells, whereas it did not alter the cellular accumulation of daunorubicin in MCF-7/sensitive cells. Similarly, the presence of macelignan also enhanced significantly (P < 0.05) the cellular accumulation of rhodamine 123 in a concentration-dependent manner in NCI/ADR-RES cells. Furthermore, cancer cells were more susceptible to the cytotoxicity of vinblastine, a P-gp substrate, in the presence of macelignan. Those results suggest that macelignan has inhibitory effects on P-gp mediated cellular efflux. However, P-gp activity did not affect the cellular accumulation of macelignan itself. Taken all together, macelignan was identified as a novel inhibitor of P-gp activity and may be a promising lead compound for the rational design of more efficacious drugs to reverse multidrug resistance in cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Lignans/pharmacology , Adenocarcinoma , Analysis of Variance , Antineoplastic Agents, Phytogenic/pharmacokinetics , Biological Transport, Active/drug effects , Breast Neoplasms , Cell Line, Tumor , Cell Survival/drug effects , Daunorubicin/pharmacokinetics , Dose-Response Relationship, Drug , Drug Design , Drug Resistance, Multiple , Drug Synergism , Female , Fluorescent Dyes/pharmacokinetics , Growth Inhibitors/pharmacokinetics , Growth Inhibitors/pharmacology , Humans , Lignans/pharmacokinetics , Phytoestrogens/pharmacokinetics , Phytoestrogens/pharmacology , Phytotherapy , Rhodamine 123/pharmacokinetics , Vinblastine/pharmacokinetics
13.
J Nat Prod ; 72(9): 1616-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19739602

ABSTRACT

Oroxylin A (1), a flavonoid from the roots of Scutellaria baicalensis, increased the cellular accumulation of calcein AM in a concentration-dependent manner in NCI/ADR-RES cells overexpressing P-glycoprotein over the concentration range 0-40 microM. In addition, 1 significantly (p < 0.05) increased the cellular accumulation of paclitaxel in NCI/ADR-RES cells while it did not alter the cellular accumulation of paclitaxel in cells lacking P-glycoprotein expression. Accordingly, the concentrations that yielded 50% cytotoxicity of vinblastine and paclitaxel were reduced by approximately 5-fold in the presence of 1. This indicated that cancer cells became more susceptible to the cytotoxicity of vinblastine and paclitaxel in the presence of 1. The concomitant use of 1 (30 mg.kg(-1)) significantly (p < 0.05) enhanced the oral exposure of paclitaxel (15 mg.kg(-1)) in rats. The C(max) and AUC values of paclitaxel increased by 2.1-2.6-fold in the presence of 1 with no significant change in T(max). In conclusion, 1 was effective in inhibiting P-glycoprotein-mediated drug efflux both in vitro and in vivo, suggesting that it may be useful to improve the cellular availability of P-glycoprotein substrates such as anticancer drugs.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Flavonoids/pharmacology , Administration, Oral , Animals , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Humans , Molecular Structure , Paclitaxel/pharmacology , Plants, Medicinal/chemistry , Rats , Scutellaria baicalensis/chemistry , Vinblastine/pharmacology
14.
J Pharm Sci ; 95(8): 1684-92, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16732590

ABSTRACT

Present study aims to improve efficiency and capacity of in vivo rat pharmacokinetic studies for rapid assessment of systemic exposure (AUC and C(max)) of new chemical entities. Plasma concentration-time profiles in rats from structurally diverse compounds were extracted from the Pfizer database. AUC(0-8) was calculated with 7 data points or a reduced subset of 3 data points. AUC values determined with 7 data points were compared to subset AUC values. A < or = 30% difference in values for 90% of cases was acceptance criteria. In parallel, samples were analyzed individually and pooled at each time point across compounds. For 96% of cases, AUC values estimated using 1, 4, and 8 h were comparable to AUC values obtained from 7 data points suggesting 1, 4, and 8 h sampling should be sufficient to estimate AUC. For C(max), the difference between 1, 4, and 8 h data-point analysis versus 7 data-point analysis is less than 30% for 72% of cases. Concentrations from individual versus pooled sample analysis were found to be equivalent. A rapid rat PK screening paradigm was created by the combination of 1, 4, and 8 h sampling and pooled sample analysis, which improves throughput and cycle time of in vivo PK studies.


Subject(s)
Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/blood , Technology, Pharmaceutical/methods , Administration, Oral , Animals , Area Under Curve , Drug Evaluation, Preclinical/methods , Rats
SELECTION OF CITATIONS
SEARCH DETAIL