Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Clin Pharmacol Ther ; 115(3): 440-451, 2024 03.
Article in English | MEDLINE | ID: mdl-38235832

ABSTRACT

Intravenous or subcutaneous routes of administration (ROAs) are common dosing routes for therapeutic proteins. Eleven therapeutic proteins with approval for one ROA have subsequently received approval for a second ROA. The clinical programs supporting the second ROA consistently leveraged data from the first ROA and included studies that characterized the pharmacokinetics (PKs) of the drug administered by the new ROA to identify an appropriate dosage regimen. The selected dosing regimen was then further evaluated in clinical trials designed with various primary end points. All programs implemented model-informed drug development approaches to ensure that the selected regimens would achieve comparable systemic exposures (PK-based bridging) or pharmacodynamic (PD) responses (PD-based bridging) as the reference ROA. To support the approval of a second ROA, these programs either demonstrated noninferiority in PK, PD, and/or clinical end points for the second ROA, or established efficacy and safety through a comparison to a placebo treatment. The accumulative examples showed that clinical trials which provided the primary evidence to support approvals of the second ROA generally demonstrated noninferiority in the systemic exposures regardless of being specified as an end point or not in the study protocols. The experience to date supports the use of PK- and PD-based bridging approaches not only in the selection of dosing regimens for a second ROA to be tested in clinical studies, but also for providing evidence of effectiveness to support approval, when appropriate.


Subject(s)
Pharmacology, Clinical , Humans , Pharmaceutical Preparations , Administration, Intravenous , Drug Approval
3.
Clin Transl Sci ; 16(8): 1323-1330, 2023 08.
Article in English | MEDLINE | ID: mdl-37157935

ABSTRACT

Recently, digital health technologies (DHTs) and digital biomarkers have gained a lot of traction in clinical investigations, motivating sponsors, investigators, and regulators to discuss and implement integrated approaches for deploying DHTs. These new tools present new and unique challenges for optimal technology integration in clinical trial processes, including operational, ethical, and regulatory issues. In this paper, we gathered different perspectives to discuss challenges and perspectives from three different stakeholders: industry, US regulators, and a public-private partnership consortium. The complexities of DHT implementation, which include regulatory definitions, defining the scope of validation experiments, and the need for partnerships between BioPharma and the technology sectors, are highlighted. Most of these challenges are related to translation of DHT-derived measures into endpoints that are meaningful to clinicians and patients, participant safety, training, and retention and privacy of data. The example of the Wearable Assessments in the Clinic and Home in PD (WATCH-PD) study is discussed as an example that demonstrated the advantages of pre-competitive collaborations, which include early regulatory feedback, data sharing, and multistakeholder alignment. Future advances in DHTs are expected to spur device-agnostic measured development and incorporate patient reported outcomes in drug development. More efforts are needed to define validation experiments for a defined context of use, incentivize data sharing and development of data standards. Multistakeholder collaborations via precompetitive consortia will help facilitate broad acceptance of DHT-enabled measures in drug development.


Subject(s)
Digital Technology , Drug Development , Humans , Information Dissemination
4.
J Pharmacokinet Pharmacodyn ; 50(6): 429-444, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37140724

ABSTRACT

Pediatric populations represent a major fraction of rare diseases and compound the intrinsic challenges of pediatric drug development and drug development for rare diseases. The intertwined complexities of pediatric and rare disease populations impose unique challenges to clinical pharmacologists and require integration of novel clinical pharmacology and quantitative tools to overcome multiple hurdles during the discovery and development of new therapies. Drug development strategies for pediatric rare diseases continue to evolve to meet the inherent challenges and produce new medicines. Advances in quantitative clinical pharmacology research have been a key component in advancing pediatric rare disease research to accelerate drug development and inform regulatory decisions. This article will discuss the evolution of the regulatory landscape in pediatric rare diseases, the challenges encountered during the design of rare disease drug development programs and will highlight the use of innovative tools and potential solutions for future development programs.


Subject(s)
Pharmacology, Clinical , Pharmacology , Physicians , Child , Humans , Rare Diseases/drug therapy , Drug Development
5.
Clin Pharmacol Ther ; 111(4): 786-798, 2022 04.
Article in English | MEDLINE | ID: mdl-34860361

ABSTRACT

Several challenges are associated with rare disease drug development in neurology. In this article, we summarize the US Food and Drug Administration's experience with clinical drug development for rare neurological diseases and discuss clinical pharmacology's critical contributions to drug development for rare diseases. We used publicly available information to identify and screen drug products approved for rare neurological indications between 1983 and 2019. We highlighted cases in which clinical pharmacology contributed to the evidence of drug efficacy, dose selection for pivotal clinical trials, dose optimization based on intrinsic and extrinsic factors, pharmacokinetic bridging for formulations, and efficacy bridging across different racial groups. Fifty-one approved drug products were identified since the introduction of the Orphan Drug Act in 1983. Interestingly, the number of approvals in the last few years increased significantly, probably due to advances in genomic research and targeted drug modalities. Evaluation of dose selection in patient populations showed that in 52% of cases, the sponsors did not evaluate efficacy for more than one or two dose levels throughout the development program. Clinical pharmacology studies to evaluate the effect of intrinsic or extrinsic factors were adequately characterized in most of the applications. With the expansion of model informed drug development applications, (e.g., quantitative systems pharmacology and deep learning neural network models), the role and impact of clinical pharmacology is expected to grow exponentially in the next decade and enhance the development of novel treatment modalities for neurological rare diseases.


Subject(s)
Nervous System Diseases , Neurology , Pharmacology, Clinical , Drug Approval , Drug Development , Humans , Nervous System Diseases/drug therapy , Orphan Drug Production , Rare Diseases/drug therapy , United States , United States Food and Drug Administration
6.
J Clin Pharmacol ; 60(11): 1441-1452, 2020 11.
Article in English | MEDLINE | ID: mdl-32453882

ABSTRACT

The development of modified-release (MR) drug products aims to address a clinical need such as improving patient compliance. There are multiple pathways and development strategies for the registration and approval of MR products. The development strategy of an MR product is usually dependent on the availability and pharmacokinetic/pharmacodynamics (PK/PD) characteristics of the reference drug product, that is, an immediate-release (IR) product or a reference MR. Compared with a reference IR product, an MR product is likely to have a different PK profile over the least common dosing time due to unequal dosing intervals. In case of differences in PK profiles between the MR product and the reference product, confirmatory efficacy and safety studies may be needed to support registration. In some cases, however, a thorough clinical PK/PD characterization may provide sufficient basis to support the approval of the proposed MR product without the need for additional safety and efficacy studies. This article summarizes the US Food and Drug Administration experience and the regulatory considerations supporting the approval of MR products in the past 6 years and discusses cases in which clinical pharmacology and PK/PD information were leveraged to support approval without the need for additional clinical studies. Details of all these cases are available in the public domain. In 2 cases a well-characterized exposure-response relationship provided sufficient justification that differences in the shape of the PK profiles were not clinically relevant. In the remaining 3 cases a thorough characterization of the PK profile along with a risk-based approach provided bases for approval.


Subject(s)
Delayed-Action Preparations/pharmacokinetics , Drug Approval/methods , Drug Development , Pharmacology, Clinical/methods , Clinical Trials as Topic , Databases, Factual , Delayed-Action Preparations/adverse effects , Dose-Response Relationship, Drug , Humans , Therapeutic Equivalency , United States , United States Food and Drug Administration
7.
Neuropharmacology ; 79: 668-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24467845

ABSTRACT

Alzheimer's disease (AD) has a characteristic hallmark of amyloid-ß (Aß) accumulation in the brain. This accumulation of Aß has been related to its faulty cerebral clearance. Indeed, preclinical studies that used mice to investigate Aß clearance showed that efflux across blood-brain barrier (BBB) and brain degradation mediate efficient Aß clearance. However, the contribution of each process to Aß clearance remains unclear. Moreover, it is still uncertain how species differences between mouse and human could affect Aß clearance. Here, a modified form of the brain efflux index method was used to estimate the contribution of BBB and brain degradation to Aß clearance from the brain of wild type mice. We estimated that 62% of intracerebrally injected (125)I-Aß40 is cleared across BBB while 38% is cleared by brain degradation. Furthermore, in vitro and in silico studies were performed to compare Aß clearance between mouse and human BBB models. Kinetic studies for Aß40 disposition in bEnd3 and hCMEC/D3 cells, representative in vitro mouse and human BBB models, respectively, demonstrated 30-fold higher rate of (125)I-Aß40 uptake and 15-fold higher rate of degradation by bEnd3 compared to hCMEC/D3 cells. Expression studies showed both cells to express different levels of P-glycoprotein and RAGE, while LRP1 levels were comparable. Finally, we established a mechanistic model, which could successfully predict cellular levels of (125)I-Aß40 and the rate of each process. Established mechanistic model suggested significantly higher rates of Aß uptake and degradation in bEnd3 cells as rationale for the observed differences in (125)I-Aß40 disposition between mouse and human BBB models. In conclusion, current study demonstrates the important role of BBB in the clearance of Aß from the brain. Moreover, it provides insight into the differences between mouse and human BBB with regards to Aß clearance and offer, for the first time, a mathematical model that describes Aß clearance across BBB.


Subject(s)
Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Endothelial Cells/metabolism , Models, Neurological , Peptide Fragments/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Brain/blood supply , Cells, Cultured , Humans , Iodine Radioisotopes , Kinetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , Mice , Mice, Inbred C57BL , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Receptors, LDL/metabolism , Species Specificity , Tumor Suppressor Proteins/metabolism
8.
Fitoterapia ; 84: 347-59, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23266736

ABSTRACT

γ-Tocotrienol and sesamin are phytochemicals that display potent anticancer activity. Since sesamin inhibits the metabolic degradation of tocotrienols, studies were conducted to determine if combined treatment with sesamin potentiates the antiproliferative effects of γ-tocotrienol on neoplastic mouse (+SA) and human (MCF-7 and MDA-MB-231) mammary cancer cells. Results showed that treatment with γ-tocotrienol or sesamin alone induced a significant dose-responsive growth inhibition, whereas combination treatment with these agents synergistically inhibited the growth of +SA, MCF-7 and MDA-MB-231 mammary cancer cells, while similar treatment doses were found to have little or no effect on normal (mouse CL-S1 and human MCF-10A) mammary epithelial cell growth or viability. However, sesamin synergistic enhancement of γ-tocotrienol-induced anticancer effects was not found to be mediated from a reduction in γ-tocotrienol metabolism. Rather, combined treatment with subeffective doses of γ-tocotrienol and sesamin was found to induce G1 cell cycle arrest, and a corresponding decrease in cyclin D1, CDK2, CDK4, CDK6, phospho-Rb, and E2F1 levels, and increase in p27 and p16 levels. Additional studies showed that the antiproliferative effect of combination treatment did not initiate apoptosis or result in a decrease in mammary cancer cell viability. Taken together, these findings indicate that the synergistic antiproliferative action of combined γ-tocotrienol and sesamin treatment in mouse and human mammary cancer cells is cytostatic, not cytotoxic, and results from G1 cell cycle arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Chromans/pharmacology , Dioxoles/pharmacology , Dioxoles/pharmacokinetics , Lignans/pharmacology , Lignans/pharmacokinetics , Vitamin E/analogs & derivatives , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Chromans/administration & dosage , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Cyclin-Dependent Kinase Inhibitor Proteins/metabolism , Cyclins/genetics , Cyclins/metabolism , Dioxoles/administration & dosage , Drug Synergism , Drug Therapy, Combination , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lignans/administration & dosage , Mice , Molecular Structure , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Vitamin E/administration & dosage , Vitamin E/pharmacology
9.
Biopharm Drug Dispos ; 33(5): 246-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22528033

ABSTRACT

The aim of this work was to compare the intestinal absorption kinetics and the bioavailability of γ-tocotrienol (γ-T3) and α-tocopherol (α-Tph) administered separately as oil solutions to rats in vivo. Also, to explain the significant difference in the oral bioavailability of the compounds: (1) the release profiles using the dynamic in vitro lipolysis model, (2) the intestinal permeability and (3) carrier-mediated uptake by Niemann-Pick C1-like 1 (NPC1L1) transporter were examined. Absolute bioavailability studies were conducted after oral administration of γ-T3 or α-Tph prepared in corn oil to rats. In situ rat intestinal perfusion with ezetimibe (a NPC1L1 inhibitor) was performed to compare intestinal permeability. The in vitro interaction kinetics with NPC1L1 was examined in NPC1L1 transfected cells. While the in vitro release studies demonstrated a significantly higher release rate of γ-T3 in the aqueous phase, the oral bioavailability of α-Tph (36%) was significantly higher than γ-T3 (9%). Consequent in situ studies revealed significantly higher intestinal permeability for α-Tph compared with γ-T3 in rats. Moreover, the NPC1L1 kinetic studies demonstrated higher Vmax and Km values for α-Tph compared with γ-T3. Collectively, these results indicate that intestinal permeability is the main contributing factor for the higher bioavailability of α-Tph. Also, these results emphasize the potentially important role of intestinal permeability in the bioavailability of γ-T3, suggesting that enhancing its permeability would increase its oral bioavailability.


Subject(s)
Intestine, Small/metabolism , alpha-Tocopherol/pharmacokinetics , gamma-Tocopherol/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Azetidines/pharmacology , Biological Availability , Corn Oil/chemistry , Drug Carriers/chemistry , Ezetimibe , In Vitro Techniques , Injections, Intravenous , Intestinal Absorption , Lipolysis/drug effects , Male , Membrane Transport Proteins/metabolism , Molecular Structure , Perfusion , Permeability , Rats , Rats, Sprague-Dawley , Solubility , alpha-Tocopherol/administration & dosage , alpha-Tocopherol/blood , gamma-Tocopherol/administration & dosage , gamma-Tocopherol/blood
10.
Mol Pharm ; 9(3): 492-504, 2012 Mar 05.
Article in English | MEDLINE | ID: mdl-22264132

ABSTRACT

The aim of this work was to extrapolate in vitro and preclinical animal data to simulate the pharmacokinetic parameters of UK-343,664, a P-glycoprotein (P-gp) and CYP3A4 substrate, in human. In addition, we aimed to develop a simulation model to demonstrate the involvement and the controversial complex interaction of intestinal P-gp and CYP3A4 in its nonlinear absorption, first-pass extraction, and pharmacokinetics using the advanced compartmental absorption and transit (ACAT) model. Finally, we aimed to compare the results predicted from the model to the reported findings in human clinical studies. In situ perfusion, allometric scaling, PBPK Rodger mechanistic approach, in vitro metabolism, and fitting to in vivo data were used to mechanistically explain the absorption, distribution and metabolism, respectively. GastroPlus was used to build the integrated simulation model in human for UK-343,664 to mechanistically explain the observed clinical data at 30, 100, 200, 400, and 800 mg oral doses. The measured in vitro value for CYP3A4 K(m) (465 µM) in rCYPs was converted to units of µg/mL, corrected for assumed microsomal binding (17.8%) and applied to all metabolic processes. The measured in vitro values of V(max) for CYP3A4 (38.9 pmol/min/pmol), 2C8, 2C9, 2C19, and 2D6 were used along with the in vitro CYP3A4 K(m) to simulate liver first pass extraction and systemic clearance. The measured in vitro values of V(max) for CYP3A4 and 2D6 were used along with the in vitro CYP3A4 K(m) to simulate gut first pass extraction. V(max) and K(m) values for P-gp were obtained by fitting to in vivo data and used to simulate gut efflux transport activity. Investigation of the interaction mechanism of P-gp and CYP3A4 in the intestine was achieved by comparing the influence of a virtual knockout of P-gp or gut metabolism on the fraction absorbed, fraction reaching the portal vein, and fraction metabolized in the gut. Comparison between simulation and in vivo results showed that the in silico simulation provided a mechanistic explanation of the observed nonlinear absorption kinetics of UK-343,664 in human following its administration in the range of 30-800 mg as oral solutions. The simulation results of the pharmacokinetic parameters, AUC and C(max), by GastroPlus were comparable with those observed in vivo. This simulation model is one possible mechanistic explanation of the observed in vivo data and suggests that the nonlinear dose dependence could be attributed to saturation of both the efflux transport by P-gp and the intestinal metabolism. However, the concentration ranges for either protein saturation did not overlap and resulted in much greater than dose proportional increases in AUC. At low doses, producing intraenterocyte concentrations below the fitted value of K(m) for P-gp, the influence of P-gp appears to be protective and results in a lower fraction of gut 3A4 metabolism. At higher doses, as P-gp becomes saturated the fraction of gut 3A4 extraction increases, and eventually at the highest doses, where 3A4 becomes saturated, the fraction of gut 3A4 extraction again decreases. Such a complex interpretation of this in vitro-in vivo extrapolation (IVIVE) is another example of the value and insight obtained by physiologically based absorption simulation.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cytochrome P-450 CYP3A/metabolism , Piperazines/metabolism , Piperazines/pharmacokinetics , Pyrimidinones/metabolism , Pyrimidinones/pharmacokinetics , Absorption , Animals , Humans , Intestinal Absorption/physiology , Kinetics , Male , Rats , Rats, Sprague-Dawley
11.
Lipids ; 47(5): 461-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22271424

ABSTRACT

γ-Tocotrienol (γ-T3), a member of the vitamin E family, has been reported to possess an anticancer activity. γ-T3 is a lipophilic compound with low oral bioavailability. Previous studies showed that γ-T3 has low intestinal permeability. Thus, we have hypothesized that enhancing γ-T3 intestinal permeability will increase its oral bioavailability. Solid lipid nanoparticles (SLN) were tested as a model formulation to enhance γ-T3 permeability and bioavailability. γ-T3 intestinal permeability was compared using in situ rat intestinal perfusion, followed by in vivo relative oral bioavailability studies. In addition, in vitro cellular uptake of γ-T3 from SLN was compared to mixed micelles (MM) in a time and concentration-dependent studies. To elucidate the uptake mechanism(s) of γ-T3 from SLN and MM the contribution of NPC1L1 carrier-mediated uptake, endocytosis and passive permeability were investigated. In situ studies demonstrated SLN has tenfold higher permeability than MM. Subsequent in vivo studies showed γ-T3 relative oral bioavailability from SLN is threefold higher. Consistent with in situ results, in vitro concentration dependent studies revealed γ-T3 uptake from SLN was twofold higher than MM. In vitro mechanistic characterization showed that while endocytosis contributes to γ-T3 uptake from both formulations, the reduced contribution of NPC1L1 to the transport of γ-T3, and passive diffusion enhancement of γ-T3 are primary explanations for its enhanced uptake from SLN. In conclusion, SLN successfully enhanced γ-T3 oral bioavailability subsequent to enhanced passive permeability.


Subject(s)
Chromans/administration & dosage , Chromans/pharmacokinetics , Intestinal Mucosa/drug effects , Lipids/chemistry , Nanoparticles/administration & dosage , Vitamin E/analogs & derivatives , Administration, Oral , Animals , Biological Availability , Intestinal Mucosa/metabolism , Lipids/administration & dosage , Lipids/pharmacology , Male , Nanoparticles/chemistry , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Vitamin E/administration & dosage , Vitamin E/pharmacokinetics
12.
Planta Med ; 77(17): 1883-9, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21870322

ABSTRACT

Echinacea species are used for beneficial effects on immune function, and various prevalent phytochemicals have immunomodulatory effects. Using a commercial E. purpurea (L.) Moench product, we have evaluated the myelopoietic effect on bone marrow of rats treated with various extracts and correlated this with their chemical class composition. Granulocyte/macrophage-colony forming cells (GM-CFCs) from femurs of female Sprague-Dawley rats were assessed at 24 h after 7 daily oral treatments. A 75% ethanolic extract at 50 mg dried weight (derived from 227 mg aerial parts) per kg body weight increased GM-CFCs by 70% but at 100 mg/kg was without effect. Ethanolic extracts from aerial parts of E. angustifolia DC. var. angustifolia and E. purpurea from the USDA North Central Regional Plant Introduction Station increased GM-CFCs by 3- and 2-fold, respectively, at 200 mg/kg (~1400 mg/kg plant material). Extract from another USDA E. angustifolia was inactive. Proton and APT NMR, MS, and TLC indicated alkylamides and caffeic-acid derivatives (CADs) present in ethanolic extracts of both the commercial and USDA-derived material. Cichoric and caftaric acids were prominent in both E. purpurea ethanolic extracts but absent in E. angustifolia. Aqueous extract of the commercial material exhibited polysaccharide and CAD signatures and was without effect on GM-CFCs. A methanol-CHCl3 fraction of commercial source, also inactive, was almost exclusively 1:4 nonanoic: decanoic acids, which were also abundant in commercial ethanolic extract but absent from USDA material. In conclusion, we have demonstrated an ethanolextractable myelostimulatory activity in Echinacea aerial parts that, when obtained from commercial herbal supplements, may be antagonized by medium-chain fatty acids presumably derived from a non-plant additive.


Subject(s)
Amides/pharmacology , Caffeic Acids/pharmacology , Echinacea/chemistry , Myelopoiesis/immunology , Plant Extracts/pharmacology , Administration, Oral , Amides/chemistry , Amides/isolation & purification , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Caffeic Acids/chemistry , Caffeic Acids/isolation & purification , Chromatography, High Pressure Liquid , Colony-Forming Units Assay , Female , Granulocytes/drug effects , Granulocytes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Medicine, Traditional , Myelopoiesis/drug effects , Plant Components, Aerial/chemistry , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plants, Medicinal/chemistry , Rats , Rats, Sprague-Dawley
13.
Biomed Chromatogr ; 25(5): 621-7, 2011 May.
Article in English | MEDLINE | ID: mdl-20734323

ABSTRACT

γ-Tocotrienol (γ-T3) is a member of the vitamin E family. Recently, γ-T3 has attracted the attention of the scientific community due to its potent anticancer activity and other therapeutic benefits. The objective of this study was to develop and validate a simple and practical reversed-phase HPLC method with satisfactory sensitivity for the routine quantification of γ-T3 in rat and human plasma. The separation of γ-T3 from the plasma components was achieved with a C(18) reversed-phase column with an isocratic elution using a mixture of methanol, ethanol and acetonitrile (85:7.5:7.5, v/v/v) with a UV detection at 295 nm. γ-T3 was extracted from rat and human plasma by liquid-liquid extraction with an average recovery of 60%. The method proved linear in the range 100-5000 ng/mL. The inter-day precision ranged from 5.8 to 12.8% and the accuracy ranged from 92.4 to 108.5%, while the intra-day precision ranged from 0.7 to 7.9% in both rat and human plasma. This data confirm that the developed method has a satisfactory sensitivity, accuracy and precision for the quantification of γ-T3 in plasma. To assess its applicability the method was successfully applied to the quantitative analysis for pharmacokinetic studies of γ-T3 in rats administered a 10 mg/kg single oral dose.


Subject(s)
Chromans/blood , Chromatography, High Pressure Liquid/methods , Chromatography, Reverse-Phase/methods , Vitamin E/analogs & derivatives , Animals , Chromans/pharmacokinetics , Drug Stability , Humans , Linear Models , Male , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Sensitivity and Specificity , Vitamin E/blood , Vitamin E/pharmacokinetics
14.
Bioorg Med Chem ; 18(22): 8066-75, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20943398

ABSTRACT

Tocotrienols are vitamin E members with potent antiproliferative activity against preneoplastic and neoplastic mammary epithelial cells with little or no effect on normal cell growth or functions. However, physicochemical and pharmacokinetic properties greatly limit their use as therapeutic agents. Tocotrienols' chemical instability, poor water solubility, NPC1L1-mediated transport, and rapid metabolism are examples of such obstacles which hinder the therapeutic use of these valuable natural products. Vitamin E esters like α-tocopheryl succinate were prepared to significantly improve chemical and metabolic stability, water solubility, and potency. Thus, 12 semisynthetic tocotrienol ester analogues 4-15 were prepared by direct esterification of natural tocotrienol isomers with various acid anhydrides or chlorides. Esters 4-15 were evaluated for their ability to inhibit the proliferation and migration of the mammary tumor cells +SA and MDA-MB-231, respectively. Esters 5, 9, and 11 effectively inhibited the proliferation of the highly metastatic +SA rodent mammary epithelial cells with IC(50) values of 0.62, 0.51, and 0.86µM, respectively, at doses that had no effect on immortalized normal mouse CL-S1 mammary epithelial cells. Esters 4, 6, 8-10, and 13 inhibited 50% of the migration of the human metastatic MDA-MB-231 breast cancer cells at a single 5µM dose in wound-healing assay. The most active ester 9 was 1000-fold more water-soluble and chemically stable versus its parent α-tocotrienol (1). These findings strongly suggest that redox-silent tocotrienol esters may provide superior therapeutic forms of tocotrienols for the control of metastatic breast cancer.


Subject(s)
Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Tocotrienols/chemistry , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Esters , Female , Humans , Mammary Glands, Animal/cytology , Mice , Oxidation-Reduction , Rats , Solubility , Tocotrienols/chemical synthesis , Tocotrienols/pharmacology
15.
Drug Metab Dispos ; 38(6): 939-45, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20207946

ABSTRACT

gamma-Tocotrienol (gamma-T3) is a member of the vitamin E family that displays potent anticancer activity and other therapeutic benefits. The objective of this study was to evaluate gamma-T3 intestinal uptake and metabolism using the in situ rat intestinal perfusion model. Isolated segments of rat jejunum and ileum were perfused with gamma-T3 solution, and measurements were made as a function of concentration (5-150 microM). Intestinal permeability (P(eff)) and metabolism were studied by measuring total compound disappearance and major metabolite, 2,7,8-trimethyl-2-(beta-carboxy-ethyl)-6-hydroxychroman, appearance in the intestinal lumen. gamma-T3 and metabolite levels were also determined in mesenteric blood. The P(eff) of gamma-T3 was similar in both intestinal segments and significantly decreased at concentrations > or =25 microM in jejunum and ileum (p < 0.05), whereas metabolite formation was minimal and mesenteric blood concentrations of gamma-T3 and metabolite remained very low. These results indicate that gamma-T3 intestinal uptake is a saturable carrier-mediated process and metabolism is minimal. Results from subsequent in situ inhibition studies with ezetimibe, a potent and selective inhibitor of Niemann-Pick C1-like 1 (NPC1L1) transporter, suggested gamma-T3 intestinal uptake is mediated by NPC1L1. Comparable findings were obtained when Madin-Darby canine kidney II cells that express endogenous NPC1L1 were incubated with increasing concentrations of gamma-T3 or gamma-T3 with increasing concentrations of ezetimibe. The present data show for the first time that gamma-T3 intestinal absorption is partly mediated by NPC1L1.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Chromans/pharmacokinetics , Intestinal Mucosa/metabolism , Jejunum/metabolism , Niemann-Pick Diseases/metabolism , Vitamin E/analogs & derivatives , Animals , Azetidines , Biological Transport , Ezetimibe , Ileum/metabolism , Intestinal Absorption/drug effects , Jejunum/drug effects , Male , Perfusion , Rats , Rats, Sprague-Dawley , Vitamin E/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...