Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Complementary Medicines
Therapeutic Methods and Therapies TCIM
Country/Region as subject
Publication year range
1.
Drug Metab Dispos ; 48(12): 1264-1270, 2020 12.
Article in English | MEDLINE | ID: mdl-33037044

ABSTRACT

Organic anion-transporting polypeptide (OATP) 1B1/3-mediated drug-drug interaction (DDI) potential is evaluated in vivo with rosuvastatin (RST) as a probe substrate in clinical studies. We calibrated our assay with RST and estradiol 17-ß-D-glucuronide (E217ßG)/cholecystokinin-8 (CCK8) as in vitro probes for qualitative and quantitative prediction of OATP1B-mediated DDI potential for RST. In vitro OATP1B1/1B3 inhibition using E217ßG and CCK8 yielded higher area under the curve (AUC) ratio (AUCR) values numerically with the static model, but all probes performed similarly from a qualitative cutoff-based prediction, as described in regulatory guidances. However, the magnitudes of DDI were not captured satisfactorily. Considering that clearance of RST is also mediated by gut breast cancer resistance protein (BCRP), inhibition of BCRP was also incorporated in the DDI prediction if the gut inhibitor concentrations were 10 × IC50 for BCRP inhibition. This combined static model closely predicted the magnitude of RST DDI with root-mean-square error values of 0.767-0.812 and 1.24-1.31 with and without BCRP inhibition, respectively, for in vitro-in vivo correlation of DDI. Physiologically based pharmacokinetic (PBPK) modeling was also used to simulate DDI between RST and rifampicin, asunaprevir, and velpatasvir. Predicted AUCR for rifampicin and asunaprevir was within 1.5-fold of that observed, whereas that for velpatasvir showed a 2-fold underprediction. Overall, the combined static model incorporating both OATP1B and BCRP inhibition provides a quick and simple mathematical approach to quantitatively predict the magnitude of transporter-mediated DDI for RST for routine application. PBPK complements the static model and provides a framework for studying molecules when a dynamic model is needed. SIGNIFICANCE STATEMENT: Using 22 drugs, we show that a static model for organic anion-transporting polypeptide (OATP) 1B1/1B3 inhibition can qualitatively predict potential for drug-drug interaction (DDI) using a cutoff-based approach, as in regulatory guidances. However, consideration of both OATP1B1/3 and gut breast cancer resistance protein inhibition provided a better prediction of the magnitude of the transporter-mediated DDI of these inhibitors with rosuvastatin. Based on these results, we have proposed an empirical mechanistic-static approach for a more reliable prediction of transporter-mediated DDI liability with rosuvastatin that drug development teams can leverage.


Subject(s)
Liver-Specific Organic Anion Transporter 1/metabolism , Models, Biological , Rosuvastatin Calcium/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Area Under Curve , Cholecystokinin/pharmacokinetics , Drug Evaluation, Preclinical , Drug Interactions , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , HEK293 Cells , Humans , Liver-Specific Organic Anion Transporter 1/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Peptide Fragments/pharmacokinetics , Solute Carrier Organic Anion Transporter Family Member 1B3/antagonists & inhibitors
2.
Theranostics ; 10(7): 3281-3292, 2020.
Article in English | MEDLINE | ID: mdl-32194868

ABSTRACT

Hormone therapy (HT) is one of the most effective treatments for osteoporosis. However, the nonselective accumulation of hormone in organs such as breast, heart and uterus other than bones causes serious side effects, which impedes the application of HT. Hence, it is critically important to develop a HT strategy with reduced non-specific enrichment of hormone drugs in non-target tissues and enhanced bone-targeting ability. Methods: Herein, a 17ß-estradiol (E2)-laden mesoporous silica-coated upconversion nanoparticle with a surface modification of ethylenediaminetetraacetic acid (EDTA) (NaLuF4:Yb,Tm@NaLuF4@mSiO2-EDTA-E2, E2-csUCNP@MSN-EDTA) is developed for bone-targeted osteoporosis hormone therapy. EDTA was attached onto the surface of E2 upconversion nanocomposite to enhance its affinity and efficiency targeting bone tissue and cells to optimize hormone replacement therapy for osteoporosis. We characterized the size, cytotoxicity, loading and release efficiency, in situ and ex vivo imaging. Further, in vitro and in vivo osteogenic ability was tested using preosteoblast and ovariectomy mouse model of osteoporosis. Results: The upconversion core of E2-csUCNP@MSN-EDTA nanoparticle serves as an excellent imaging agent for tracking the loaded hormone drug in vivo. The mesoporous silica layer has a high loading efficiency for E2 and provides a relatively long-lasting drug release within 50 h. EDTA anchored on the silica layer endows the nanocomposite with a bone targeting property. The nanocomposite effectively reverses estrogen deficiency-induced osteoporosis and reduces the damage of hormone to the uterus. The bone mineral density in the nanocomposite treatment group is nearly twice that of the ovariectomized (OVX) group. Compared with the E2 group, the uterine weight and luminal epithelial height were significantly lower in the nanocomposite treatment group. Conclusion: This work demonstrated that E2-csUCNP@MSN-EDTA alleviates the side effect of hormone therapy while maintaining its therapeutic efficacy, which has great potential for developing the next generation of methods for osteoporosis treatment.


Subject(s)
Edetic Acid/administration & dosage , Estradiol/administration & dosage , Hormone Replacement Therapy/methods , Nanocomposites/administration & dosage , Nanoparticles/administration & dosage , Osteoporosis/drug therapy , Animals , Cell Line , Edetic Acid/pharmacokinetics , Edetic Acid/toxicity , Estradiol/pharmacokinetics , Estradiol/therapeutic use , Estradiol/toxicity , Female , Mice , Nanocomposites/toxicity , Nanoparticles/toxicity , Organ Specificity , Osteoblasts/drug effects , Ovariectomy , Tissue Distribution , Uterus/drug effects , Whole Body Imaging
3.
Drug Metab Dispos ; 48(10): 966-971, 2020 10.
Article in English | MEDLINE | ID: mdl-31900255

ABSTRACT

Curcumin, a major polyphenol present in turmeric, is predominantly converted to curcumin-O-glucuronide (COG) in enterocytes and hepatocytes via glucuronidation. COG is a principal metabolite of curcumin in plasma and feces. It appears that the efflux transport of the glucuronide conjugates of many compounds is mediated largely by multidrug resistance-associated protein (MRP) 3, the gene product of the ATP-binding cassette, subfamily C, member 3. However, it is currently unknown whether this was the case with COG. In this study, Mrp3 knockout (KO) and wild-type (WT) mice were used to evaluate the pharmacokinetics profiles of COG, the liver-to-plasma ratio of COG, and the COG-to-curcumin ratio in plasma, respectively. The ATP-dependent uptake of COG into recombinant human MRP3 inside-out membrane vesicles was measured for further identification, with estradiol-17ß-d-glucuronide used in parallel as the positive control. Results showed that plasma COG concentrations were extremely low in KO mice compared with WT mice, that the liver-to-plasma ratios of COG were 8-fold greater in KO mice than in WT mice, and that the ATP-dependent uptake of COG at 1 or 10 µM was 5.0- and 3.1-fold greater in the presence of ATP than in the presence of AMP, respectively. No significant differences in the Abcc2 and Abcg2 mRNA expression levels were seen between Mrp3 KO and WT mice. We conclude that Mrp3 is identified to be the main efflux transporter responsible for the transport of COG from hepatocytes into the blood. SIGNIFICANCE STATEMENT: This study was designed to determine whether multidrug resistance-associated protein (Mrp) 3 could be responsible for the efflux transport of curcumin-O-glucuronide (COG), a major metabolite of curcumin present in plasma and feces, from hepatocytes into the blood using Mrp3 knockout mice. In this study, COG was identified as a typical Mrp3 substrate. Results suggest that herb-drug interactions would occur in patients concomitantly taking curcumin and either an MRP3 substrate/inhibitor or a drug that is predominantly glucuronidated by UDP-glucuronosyltransferases.


Subject(s)
Curcumin/analogs & derivatives , Glucuronides/pharmacokinetics , Hepatocytes/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Administration, Oral , Animals , Curcumin/administration & dosage , Curcumin/pharmacokinetics , Drug Evaluation, Preclinical , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacokinetics , Glucuronides/administration & dosage , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics
4.
Hypertension ; 73(5): 1128-1136, 2019 05.
Article in English | MEDLINE | ID: mdl-30929518

ABSTRACT

Low birth weight is associated with a greater prevalence of hypertension and an earlier age at menopause in women in later life. Yet, the association between birth weight and blood pressure (BP) in women as they age is not well defined. In a rodent model of low birth weight induced by placental insufficiency, intrauterine growth restriction programs a significant increase in BP by 12 months of age in female growth-restricted offspring that is associated with early reproductive senescence, increased testosterone, and a shift in the hormonal milieu. Thus, this study tested the hypothesis that increased BP in female growth-restricted offspring is abolished by chronic estradiol supplementation. Placebo or 17ß-estradiol valerate mini pellets (1.5 mg for 60-day release) were administered at 12 months of age for 6 weeks. BP, measured in conscious catheterized rats, was significantly increased in placebo-treated growth-restricted relative to placebo-treated control. However, BP was not elevated in estradiol-treated growth-restricted relative to placebo-treated growth-restricted. Estradiol mediates its effects on BP via its receptors and the renin-angiotensin system. BP was decreased in growth-restricted offspring treated with a G-protein coupled receptor agonist, G1 (400 mg/kg for 2 weeks). Renal AT1aR (angiotensin type 1a receptor) and AT1bR (angiotensin type 1b receptor) and renal AR (androgen receptor) mRNA expression were elevated in vehicle-treated growth-restricted offspring, but not in G1-treated growth-restricted. Therefore, these data suggest that chronic estradiol supplementation prevents the increase in BP that develops in female growth-restricted offspring via actions that may involve its G-protein coupled receptor and the renin-angiotensin system.


Subject(s)
Blood Pressure/drug effects , Estradiol/administration & dosage , Fetal Growth Retardation/prevention & control , Pregnancy, Animal , Animals , Dietary Supplements , Disease Models, Animal , Dose-Response Relationship, Drug , Estradiol/pharmacokinetics , Estrogens/administration & dosage , Estrogens/pharmacokinetics , Female , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/physiopathology , Male , Pregnancy , Rats
5.
Drug Metab Pharmacokinet ; 30(5): 374-83, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26403084

ABSTRACT

Berberine is a pharmacologically active alkaloid present in widely used medicinal plants, such as Coptis chinensis (Huang-Lian). The hormone estradiol is oxidized by cytochrome P450 (CYP) 1B1 to primarily form the genotoxic metabolite 4-hydroxyestradiol, whereas CYP1A1 and CYP1A2 predominantly generate 2-hydroxyestradiol. To illustrate the effect of berberine on the regioselective oxidation of estradiol, effects of berberine and its metabolites on CYP1 activities were studied. Among CYP1s, CYP1B1.1, 1.3 (L432V), and 1.4 (N453S)-catalyzed 4-hydroxylation were preferentially inhibited by berberine. Differing from the competitive inhibition of CYP1B1.1 and 1.3, N453S substitution in CYP1B1 allowed a non-competitive or mixed-type pattern. An N228T in CYP1B1 highly decreased its activity and preference to 4-hydroxylation. A reverse mutation of T223N in CYP1A2 retained its 2-hydroxylation preference, but enhanced its inhibition susceptibility to berberine. Compared with berberine, metabolites demethyleneberberine and thalifendine caused weaker inhibition of CYP1A1 and CYP1B1 activities. Unexpectedly, thalifendine was more potent than berberine in the inhibition of CYP1A2, in which case an enhanced interaction through polar hydrogen-π bond was predicted from the docking analysis. These results demonstrate that berberine preferentially inhibits the estradiol 4-hydroxylation activity of CYP1B1 variants, suggesting that 4-hydroxyestradiol-mediated toxicity might be reduced by berberine, especially in tissues/tumors highly expressing CYP1B1.


Subject(s)
Berberine/pharmacology , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Estradiol/pharmacokinetics , Berberine/analogs & derivatives , Berberine/metabolism , Berberine/pharmacokinetics , Computer Simulation , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Humans , Hydroxylation , Isoenzymes/genetics , Isoenzymes/metabolism , Molecular Docking Simulation , Oxidation-Reduction , Recombinant Proteins/metabolism
6.
J Control Release ; 180: 117-24, 2014 Apr 28.
Article in English | MEDLINE | ID: mdl-24556419

ABSTRACT

The primary objective of this study was to compare the biodistribution and pharmacokinetic profile of 17-ß-estradiol (17-ßE) on systemic delivery using either the cationic or the CREKA-peptide-modified (Cysteine-Arginine-Glutamic-acid-Lysine-Alanine) omega-3-fatty acid oil containing nanoemulsion system in vivo in the wild type C57BL/6 mice. Higher blood concentrations of 17-ßE, higher accumulation in the tissues of interest - heart and aorta, and higher accumulation within the other tissues - liver and kidney was observed on delivering 17-ßE using the CREKA-peptide-modified nanoemulsion system (AUClast in plasma - 263.89±21.81min*%/injected dose/ml) as compared to the cationic nanoemulsion (AUClast in plasma - 20.2±1.86min*%/injected dose/ml) and solution form (AUClast in plasma - 44.9±1.24min*%/injected dose/ml) respectively. Both, the cationic nanoemulsion and the CREKA-peptide-modified nanoemulsion showed a higher relative targeting efficiency of 4.57 and 4.86 respectively for 17-ßE than the relative targeting efficiency of 1.78 observed with the solution form. In conclusion, since the maximum exposure (highest AUClast for plasma and tissues) for 17-ßE was observed with the CREKA-peptide-modified nanoemulsion system, the study shows that CREKA-peptide-modified nanoemulsion system was the most suitable vehicle for systemic delivery of 17-ßE in the wild type C57BL/6 mice.


Subject(s)
Drug Carriers/chemistry , Emulsions/chemistry , Estradiol/administration & dosage , Estradiol/pharmacokinetics , Fatty Acids, Omega-3/chemistry , Oligopeptides/chemistry , Animals , Atherosclerosis/drug therapy , Cations/chemistry , Estradiol/blood , Estradiol/metabolism , Male , Mice , Mice, Inbred C57BL , Tissue Distribution
7.
Eur J Endocrinol ; 170(1): 121-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24114435

ABSTRACT

OBJECTIVE: Estrogen stimulates pulsatile secretion of GH, via mechanisms that are largely unknown. An untested hypothesis is that estradiol (E2) drives GH secretion by amplifying interactions among GH-releasing hormone (GHRH), somatostatin (SS), and GH-releasing peptide (GHRP). DESIGN: The design comprised double-blind randomized prospective administration of transdermal E2 vs placebo to healthy postmenopausal women (n=24) followed by pulsatile GHRH or SS infusions for 13 h overnight with or without continuous GHRP2 stimulation. METHODS: End points were mean concentrations, deconvolved secretion, and approximate entropy (ApEn; a regularity measure) of GH. RESULTS: By generalized ANOVA models, it was observed that E2 vs placebo supplementation: i) augmented mean (13-h) GH concentrations (P=0.023), GHRH-induced pulsatile GH secretion over the first 3 h (P=0.0085) and pulsatile GH secretion over the next 10 h (P=0.054); ii) increased GHRP-modulated (P=0.022) and SS-modulated (P<0.001) GH ApEn; and iii) did not amplify GHRH/GHRP synergy during pulsatile GH secretion. By linear regression, E2 concentrations were found to be positively correlated with GH secretion during GHRP2 infusion (P=0.022), whereas BMI was found to be negatively correlated with GH secretion during GHRH (P=0.006) and combined GHRH/GHRP (P=0.015) stimulation. E2 and BMI jointly determined triple (combined l-arginine, GHRH, and GHRP2) stimulation of GH secretion after saline (R²=0.44 and P=0.003) and pulsatile GHRH (R²=0.39 and P=0.013) infusions. CONCLUSION: In summary, in postmenopausal women, E2 supplementation augments the amount (mass) and alters the pattern (regularity) of GH secretion via interactions among GHRH, SS, GHRP, and BMI. These outcomes introduce a more complex model of E2 supplementation in coordinating GH secretion in aging women.


Subject(s)
Estradiol/therapeutic use , Estrogen Replacement Therapy , Growth Hormone-Releasing Hormone/metabolism , Human Growth Hormone/metabolism , Overweight/metabolism , Pituitary Gland, Anterior/drug effects , Somatostatin/metabolism , Aged , Arginine/administration & dosage , Body Mass Index , Double-Blind Method , Estradiol/administration & dosage , Estradiol/adverse effects , Estradiol/pharmacokinetics , Estrogen Replacement Therapy/adverse effects , Feedback, Physiological/drug effects , Female , Growth Hormone-Releasing Hormone/administration & dosage , Human Growth Hormone/blood , Humans , Infusions, Intravenous , Middle Aged , Oligopeptides/administration & dosage , Oligopeptides/metabolism , Overweight/blood , Overweight/physiopathology , Pituitary Gland, Anterior/metabolism , Pituitary Gland, Anterior/physiopathology , Postmenopause , Pulsatile Flow/drug effects , Somatostatin/administration & dosage , Transdermal Patch
8.
J Reprod Dev ; 59(6): 563-8, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24047956

ABSTRACT

The aim of the present study was to determine if the estradiol-induced luteinizing hormone (LH) surge is influenced by the constant exposure to TAK-683, an investigational metastin/kisspeptin analog, that had been established to depress the pulsatile gonadotropin-releasing hormone (GnRH) and LH secretion in goats. Ovariectomized goats subcutaneously received TAK-683 (TAK-683 group, n=6) or vehicle (control group, n=6) constantly via subcutaneous implantation of an osmotic pump. Five days after the start of the treatment, estradiol was infused intravenously in both groups to evaluate the effects on the LH surge. Blood samples were collected at 6-min intervals for 4 h prior to the initiation of either the TAK-683 treatment or the estradiol infusion, to determine the profiles of pulsatile LH secretion. They were also collected at 2-h intervals from -4 h to 32 h after the start of estradiol infusion for analysis of LH surges. The frequency and mean concentrations of LH pulses in the TAK-683 group were remarkably suppressed 5 days after the start of TAK-683 treatment compared with those of the control group (P<0.05). On the other hand, a clear LH surge was observed in all animals of both groups. There were no significant differences in the LH concentrations for surge peak and the peak time of the LH surge between the TAK-683 and control groups. These findings suggest that the effects of continuous exposure to kisspeptin or its analog on the mechanism(s) that regulates the pulsatile and surge mode secretion of GnRH/LH are different in goats.


Subject(s)
Drugs, Investigational/administration & dosage , Hypothalamus/drug effects , Kisspeptins/administration & dosage , Luteinizing Hormone/metabolism , Neurons/drug effects , Receptors, G-Protein-Coupled/agonists , Secretory Pathway/drug effects , Animals , Animals, Inbred Strains , Anterior Hypothalamic Nucleus/drug effects , Anterior Hypothalamic Nucleus/metabolism , Drug Implants , Drugs, Investigational/pharmacology , Estradiol/blood , Estradiol/pharmacokinetics , Estradiol/pharmacology , Female , Goats , Hypothalamus/metabolism , Infusions, Subcutaneous , Japan , Jugular Veins , Kisspeptins/pharmacology , Luteinizing Hormone/blood , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Ovariectomy , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/drug effects , Preoptic Area/metabolism , Receptors, G-Protein-Coupled/metabolism , Secretory Rate/drug effects
9.
Antimicrob Agents Chemother ; 57(8): 3994-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23752507

ABSTRACT

Multipurpose technologies that simultaneously protect from sexually transmitted infections and unintended pregnancy are urgently needed. Pod-intravaginal rings (IVRs) formulated with the antiretroviral agents (ARVs) tenofovir, nevirapine, and saquinavir and the contraceptives etonogestrel and estradiol were evaluated in sheep. Steady-state concentrations were maintained for 28 days with controlled, sustained delivery. This proof-of-principle study demonstrates that pod IVRs can deliver three ARVs from different mechanistic classes and a progestin-estrogen combination over the wide range needed for putative preventative efficacy.


Subject(s)
Anti-Retroviral Agents/pharmacokinetics , Contraceptive Agents/pharmacokinetics , Intrauterine Devices, Medicated , Adenine/administration & dosage , Adenine/analogs & derivatives , Adenine/pharmacokinetics , Administration, Intravaginal , Animals , Anti-Retroviral Agents/administration & dosage , Biopsy , Contraceptive Agents/administration & dosage , Desogestrel/administration & dosage , Desogestrel/pharmacokinetics , Drug Evaluation, Preclinical , Estradiol/administration & dosage , Estradiol/pharmacokinetics , Female , Models, Animal , Nevirapine/administration & dosage , Nevirapine/pharmacokinetics , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Saquinavir/administration & dosage , Saquinavir/pharmacokinetics , Sexually Transmitted Diseases, Viral/prevention & control , Sheep , Tenofovir , Time Factors
10.
Expert Rev Anticancer Ther ; 11(11): 1641-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22050013

ABSTRACT

Fulvestrant is a form of endocrine therapy used in the treatment of postmenopausal breast cancer. It has a unique mechanism of action in that it causes the degradation of estrogen receptor and therefore has been labeled a selective estrogen receptor downregulator. Unlike the selective estrogen receptor modulator tamoxifen, it has no agonistic properties and is therefore a pure anti-estrogen. Given its low level of bioavailability and presystemic metabolism, it has been formulated as an intramuscular injection. A number of dosing regimens have been utilized - these include a dose of 250 mg monthly ('approved dose'), an initial 500 mg followed by 250 mg on days 14 and 28, and thereafter 250 mg every 28 days ('loading dose'), or 500 mg on days 0, 14 and 28, and thereafter every 28 days ('high dose'). This article will review its unique mode of action and preclinical data, as well as clinical data for different dosing regimens and data for its combination with aromatase inhibitors. Fulvestrant is a well-tolerated drug and its toxicities will also be reviewed. The optimal position of fulvestrant in sequential endocrine therapy has yet to be defined.


Subject(s)
Breast Neoplasms/drug therapy , Estradiol/analogs & derivatives , Postmenopause/metabolism , Animals , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/metabolism , Clinical Trials as Topic , Drug Evaluation, Preclinical , Estradiol/pharmacokinetics , Estradiol/therapeutic use , Estrogen Antagonists/therapeutic use , Female , Fulvestrant , Humans , Receptors, Estrogen/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL