Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 336
1.
Mol Pharm ; 18(9): 3401-3417, 2021 09 06.
Article En | MEDLINE | ID: mdl-34482696

The need of pharmacological strategies to preclude breast cancer development motivated us to develop a non-aqueous microemulsion (ME) capable of forming a depot after administration in the mammary tissue and uptake of interstitial fluids for prolonged release of the retinoid fenretinide. The selected ME was composed of phosphatidylcholine/tricaprylin/propylene glycol (45:5:50, w/w/w) and presented a droplet diameter of 175.3 ± 8.9 nm. Upon water uptake, the ME transformed successively into a lamellar phase, gel, and a lamellar phase-containing emulsion in vitro as the water content increased and released 30% of fenretinide in vitro after 9 days. Consistent with the slow release, the ME formed a depot in cell cultures and increased fenretinide IC50 values by 68.3- and 13.2-fold in MCF-7 and T-47D cells compared to a solution, respectively. At non-cytotoxic concentrations, the ME reduced T-47D cell migration by 75.9% and spheroid growth, resulting in ∼30% smaller structures. The depot formed in vivo prolonged a fluorochrome release for 30 days without producing any sings of local irritation. In a preclinical model of chemically induced carcinogenesis, ME administration every 3 weeks for 3 months significantly reduced (4.7-fold) the incidence of breast tumors and increased type II collagen expression, which might contribute to limit spreading. These promising results support the potential ME applicability as a preventive therapy of breast cancer.


Anticarcinogenic Agents/administration & dosage , Breast Neoplasms/prevention & control , Fenretinide/administration & dosage , Mammary Neoplasms, Experimental/prevention & control , Animals , Anticarcinogenic Agents/pharmacokinetics , Breast Neoplasms/chemically induced , Breast Neoplasms/pathology , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Liberation , Drug Screening Assays, Antitumor , Emulsions , Female , Fenretinide/pharmacokinetics , Humans , Inhibitory Concentration 50 , MCF-7 Cells , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Methylnitrosourea/administration & dosage , Methylnitrosourea/toxicity , Mice , Rats
2.
Drug Metab Dispos ; 49(8): 694-705, 2021 08.
Article En | MEDLINE | ID: mdl-34035125

3,3'-Diindolylmethane (DIM), a major phytochemical derived from ingestion of cruciferous vegetables, is also a dietary supplement. In preclinical models, DIM is an effective cancer chemopreventive agent and has been studied in a number of clinical trials. Previous pharmacokinetic studies in preclinical and clinical models have not reported DIM metabolites in plasma or urine after oral dosing, and the pharmacological actions of DIM on target tissues is assumed to be solely via the parent compound. Seven subjects (6 males and 1 female) ranging from 26-65 years of age, on a cruciferous vegetable-restricted diet prior to and during the study, took 2 BioResponse DIM 150-mg capsules (45.3 mg DIM/capsule) every evening for one week with a final dose the morning of the first blood draw. A complete time course was performed with plasma and urine collected over 48 hours and analyzed by UPLC-MS/MS. In addition to parent DIM, two monohydroxylated metabolites and 1 dihydroxylated metabolite, along with their sulfate and glucuronide conjugates, were present in both plasma and urine. Results reported here are indicative of significant phase 1 and phase 2 metabolism and differ from previous pharmacokinetic studies in rodents and humans, which reported only parent DIM present after oral administration. 3-((1H-indole-3-yl)methyl)indolin-2-one, identified as one of the monohydroxylated products, exhibited greater potency and efficacy as an aryl hydrocarbon receptor agonist when tested in a xenobiotic response element-luciferase reporter assay using Hepa1 cells. In addition to competitive phytochemical-drug adverse reactions, additional metabolites may exhibit pharmacological activity highlighting the importance of further characterization of DIM metabolism in humans. SIGNIFICANCE STATEMENT: 3,3'-Diindolylmethane (DIM), derived from indole-3-carbinol in cruciferous vegetables, is an effective cancer chemopreventive agent in preclinical models and a popular dietary supplement currently in clinical trials. Pharmacokinetic studies to date have found little or no metabolites of DIM in plasma or urine. In marked contrast, we demonstrate rapid appearance of mono- and dihydroxylated metabolites in human plasma and urine as well as their sulfate and glucuronide conjugates. The 3-((1H-indole-3-yl)methyl)indolin-2-one metabolite exhibited significant aryl hydrocarbon receptor agonist activity, emphasizing the need for further characterization of the pharmacological properties of DIM metabolites.


Indoles , Administration, Oral , Anticarcinogenic Agents/blood , Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/urine , Capsules , Dietary Supplements , Drug Development , Drug Elimination Routes , Female , Humans , Inactivation, Metabolic/physiology , Indoles/blood , Indoles/pharmacokinetics , Indoles/urine , Male , Middle Aged , Phytochemicals/blood , Phytochemicals/pharmacokinetics , Phytochemicals/urine
3.
Molecules ; 26(2)2021 Jan 11.
Article En | MEDLINE | ID: mdl-33440795

Melatonin is a hormone secreted in the pineal gland with several functions, especially regulation of circadian sleep cycle and the biological processes related to it. This review evaluates the bioavailability of melatonin and resulting metabolites, the presence of melatonin in wine and beer and factors that influence it, and finally the different benefits related to treatment with melatonin. When administered orally, melatonin is mainly absorbed in the rectum and the ileum; it has a half-life of about 0.45-1 h and is extensively inactivated in the liver by phase 2 enzymes. Melatonin (MEL) concentration varies from picograms to ng/mL in fermented beverages such as wine and beer, depending on the fermentation process. These low quantities, within a dietary intake, are enough to reach significant plasma concentrations of melatonin, and are thus able to exert beneficial effects. Melatonin has demonstrated antioxidant, anticarcinogenic, immunomodulatory and neuroprotective actions. These benefits are related to its free radical scavenging properties as well and the direct interaction with melatonin receptors, which are involved in complex intracellular signaling pathways, including inhibition of angiogenesis and cell proliferation, among others. In the present review, the current evidence on the effects of melatonin on different pathophysiological conditions is also discussed.


Beer/analysis , Melatonin/analysis , Wine/analysis , Animals , Anticarcinogenic Agents/analysis , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/pharmacology , Antioxidants/analysis , Antioxidants/metabolism , Antioxidants/pharmacokinetics , Antioxidants/pharmacology , Circadian Rhythm/drug effects , Fermentation , Humans , Melatonin/metabolism , Melatonin/pharmacokinetics , Melatonin/pharmacology , Neuroprotective Agents/analysis , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology
4.
Biomed Pharmacother ; 134: 111124, 2021 Feb.
Article En | MEDLINE | ID: mdl-33434782

Triple-negative breast cancer (TNBC) accounts for about 15 % of all breast cancer cases, and unlike other malignancies, it lacks definite prognostic markers. While improved survival responses have been documented with the ongoing therapeutic approaches, the development of tumor resistance mechanisms to these treatment options pose major challenges in the treatment of TNBC. Notably, naturally occurring medicinal compounds have been studied extensively for their anti-neoplastic activities in cancer models including breast cancer due to their safe and non-deleterious effects. Among various dietary compounds, Withaferin-A (WA), a phytochemical derived from an ayurvedic medicinal plant, Withania somnifera has been characterized to possess anti-inflammatory and anti-cancer properties. Importantly, multiple studies have shown that WA exhibits promising anti-tumoral activities against in-vitro and in-vivo experimental models of TNBC and that its combination has been documented to enhance chemotherapy efficacy. The current review highlights the mechanistic insights with recent updates including the pharmacokinetics parameters and implications of WA against breast cancer with major emphasis on TNBC.


Anticarcinogenic Agents/therapeutic use , Cell Transformation, Neoplastic/drug effects , Triple Negative Breast Neoplasms/prevention & control , Withanolides/therapeutic use , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/pharmacokinetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Withanolides/adverse effects , Withanolides/pharmacokinetics
5.
Nutr Res ; 81: 19-37, 2020 09.
Article En | MEDLINE | ID: mdl-32828967

Chlorophyll, a phytochemical responsible for the green pigmentation in plants, has been studied for almost 100 years for its biological activities in humans. Over the past 30 years, the potential chemopreventative activities of both natural chlorophylls and their processed induced derivatives as well as the semisynthetic forms, such as sodium copper chlorophyllin, have been the focus of many research efforts. Established as potential chemopreventative agents with little to no bioavailability themselves, the activities of chlorophyll derivatives were generally ascribed to their ability to modulate mutagen/carcinogen bioavailability, their metabolism, and ultimately their ability to decrease the "exposure" to these carcinogens for humans at risk. More recently, systemic activities of chlorophyll derivatives have been reported to include modulation of oxidative stress and regulation of xenobiotic metabolizing systems and gene expression of systems critical to prevention of initiation and/or progression of cancer including NFE2-related factor 2, nuclear factor kappa B, TGF-ß, and ß-catenin pathways. With this in mind, the goals of this review are to provide an update to the comprehensive review of Ferruzzi and Blakeslee (2007) to include new insights into the behavior of chlorophyll derivatives in the gut as well as evidence of the systemic bioavailability of chlorophyll derivatives and their metabolites in support of potential impacts in prevention of cancer throughout the body.


Anticarcinogenic Agents/administration & dosage , Chlorophyll/analogs & derivatives , Diet , Neoplasms/prevention & control , Animals , Anticarcinogenic Agents/metabolism , Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/pharmacology , Biological Availability , Carcinogens/metabolism , Carcinogens/pharmacokinetics , Chemoprevention , Chlorophyll/administration & dosage , Chlorophyll/metabolism , Chlorophyll/pharmacokinetics , Digestion , Digestive System/metabolism , Humans , Intestinal Absorption , Mutagens/metabolism , Oxidative Stress , Signal Transduction , Xenobiotics/metabolism
6.
Int J Mol Sci ; 21(16)2020 Aug 10.
Article En | MEDLINE | ID: mdl-32784973

Breast cancer (BC) is the most common malignancy and the leading cause of cancer-related death in adult women worldwide. Over 85% of BC cases are non-hereditary, caused by modifiable extrinsic factors related to lifestyle, including dietary habits, which play a crucial role in cancer prevention. Although many epidemiological and observational studies have inversely correlated the fruit and vegetable consumption with the BC incidence, the involvement of their phenolic content in this correlation remains contradictory. During decades, wrong approaches that did not consider the bioavailability, metabolism, and breast tissue distribution of dietary phenolics persist behind the large currently existing gap between preclinical and clinical research. In the present review, we provide comprehensive preclinical and clinical evidence according to physiologically relevant in vitro and in vivo studies. Some dietary phenolics such as resveratrol (RSV), quercetin, isoflavones, epigallocatechin gallate (EGCG), lignans, and curcumin are gaining attention for their chemopreventive properties in preclinical research. However, the clinical evidence of dietary phenolics as BC chemopreventive compounds is still inconclusive. Therefore, the only way to validate promising preclinical results is to conduct clinical trials in BC patients. In this regard, future perspectives on dietary phenolics and BC research are also critically discussed.


Anticarcinogenic Agents/therapeutic use , Breast Neoplasms/prevention & control , Chemoprevention/methods , Dietary Supplements , Flavonoids/therapeutic use , Phenols/therapeutic use , Animals , Anticarcinogenic Agents/pharmacokinetics , Biological Availability , Breast Neoplasms/epidemiology , Clinical Trials as Topic , Diet , Disease Models, Animal , Female , Flavonoids/pharmacokinetics , Humans , Incidence , Phenols/pharmacokinetics
7.
Gastroenterology ; 159(3): 969-983.e4, 2020 09.
Article En | MEDLINE | ID: mdl-32387495

BACKGROUND & AIMS: Alterations in the intestinal microbiota affect development of colorectal cancer and drug metabolism. We studied whether the intestinal microbiota affect the ability of aspirin to reduce colon tumor development in mice. METHODS: We performed studies with APCmin/+ mice and mice given azoxymethane and dextran sulfate sodium to induce colorectal carcinogenesis. Some mice were given antibiotics to deplete intestinal microbes, with or without aspirin, throughout the entire experiment. Germ-free mice were studied in validation experiments. Colon tissues were collected and analyzed by histopathology, quantitative reverse-transcription polymerase chain reaction, and immunoblots. Blood samples and gut luminal contents were analyzed by liquid chromatography/mass spectrometry and an arylesterase activity assay. Fecal samples were analyzed by 16S ribosomal RNA gene and shotgun metagenome sequencing. RESULTS: Administration of aspirin to mice reduced colorectal tumor number and load in APCmin/+ mice and mice given azoxymethane and dextran sulfate sodium that had been given antibiotics (depleted gut microbiota), but not in mice with intact microbiota. Germ-free mice given aspirin developed fewer colorectal tumors than conventionalized germ-free mice given aspirin. Plasma levels of aspirin were higher in mice given antibiotics than in mice with intact gut microbiota. Analyses of luminal contents revealed that aerobic gut microbes, including Lysinibacillus sphaericus, degrade aspirin. Germ-free mice fed L sphaericus had lower plasma levels of aspirin than germ-free mice that were not fed this bacterium. There was an inverse correlation between aspirin dose and colorectal tumor development in conventional mice, but this correlation was lost with increased abundance of L sphaericus. Fecal samples from mice fed aspirin were enriched in Bifidobacterium and Lactobacillus genera, which are considered beneficial, and had reductions in Alistipes finegoldii and Bacteroides fragili, which are considered pathogenic. CONCLUSIONS: Aspirin reduces development of colorectal tumors in APCmin/+ mice and mice given azoxymethane and dextran sulfate sodium, depending on the presence of intestinal microbes. L sphaericus in the gut degrades aspirin and reduced its chemopreventive effects in mice. Fecal samples from mice fed aspirin were enriched in beneficial bacteria, with reductions in pathogenic bacteria.


Anticarcinogenic Agents/pharmacokinetics , Aspirin/pharmacokinetics , Colorectal Neoplasms/prevention & control , Gastrointestinal Microbiome/physiology , Adenomatous Polyposis Coli Protein/genetics , Animals , Anti-Bacterial Agents/adverse effects , Anticarcinogenic Agents/administration & dosage , Aspirin/administration & dosage , Azoxymethane/toxicity , Bacillaceae/genetics , Bacillaceae/isolation & purification , Bacillaceae/metabolism , Bacteroides fragilis/genetics , Bacteroides fragilis/isolation & purification , Bacteroides fragilis/metabolism , Bacteroidetes/genetics , Bacteroidetes/isolation & purification , Bacteroidetes/metabolism , Biological Availability , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Colitis/chemically induced , Colitis/genetics , Colon/drug effects , Colon/metabolism , Colon/microbiology , Colon/pathology , Colorectal Neoplasms/etiology , Colorectal Neoplasms/pathology , DNA, Bacterial/isolation & purification , Dextran Sulfate/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Male , Mice , Mice, Transgenic , RNA, Ribosomal, 16S/genetics
8.
Food Funct ; 11(4): 3213-3226, 2020 Apr 01.
Article En | MEDLINE | ID: mdl-32215382

Naturally occurring bioactive food components such as dietary polyphenols have shown many beneficial biological activities due to their good antioxidant properties. Among them significant attention has been given to resveratrol (RV) in recent years as it plays a promising role in cancer prevention. It has demonstrated anti-proliferative effects, as well as the ability to inhibit the initiation and progression of induced cancer in a wide variety of tumor models. However, the benefits of its therapeutic effects were found to be limited due to its poor pharmacokinetic properties such as poor aqueous solubility, instability and extensive first pass metabolism. To overcome these limitations, the present study aimed to synthesize thermosensitive copolymeric nanoparticle encapsulated formulations of resveratrol-nanoresveratrol (NRV) and evaluate their in vitro anticancer activity and inhibitory effect on 12-O-tetradecanoylphorbol-13-acetate (TPA)-promoted skin inflammation and tumorigenesis in Swiss albino mice. For this purpose PNIPAAM-PEG based thermosensitive copolymeric nanoparticles were synthesized followed by the encapsulation of RV in their hydrophobic core. This enhanced the therapeutic bioavailability of resveratrol. Nanoresveratrol demonstrated stronger antioxidant activity and comparable anticancer efficacy to free resveratrol. Nanoparticles were characterized by IR, NMR, DLS and TEM. The best results were obtained with NRV at significantly lower doses. NRV demonstrated better in vitro anticancer activity against melanoma cell line B16. It showed comparable reduction of TPA induced skin edema, hyperplasia and oxidative stress response. In the promotion phase, a significant reduction was found in tumor incidence and tumor burden in mice pre-treated with NRV. Moreover, at all doses NRV altered Bax and Bcl2 expressions which lead to the induction of apoptosis.


Anticarcinogenic Agents/pharmacokinetics , Nanoparticles/chemistry , Phytochemicals/pharmacokinetics , Resveratrol/pharmacokinetics , Animals , Anticarcinogenic Agents/analysis , Antioxidants/analysis , Antioxidants/pharmacokinetics , Apoptosis/drug effects , Biological Availability , Carcinogenesis/drug effects , Inflammation/chemically induced , Inflammation/drug therapy , Mice , Oxidative Stress/drug effects , Phytochemicals/analysis , Resveratrol/analysis , Skin/drug effects , Skin/metabolism , Tetradecanoylphorbol Acetate/toxicity
9.
J Pharm Biomed Anal ; 185: 113216, 2020 Jun 05.
Article En | MEDLINE | ID: mdl-32155543

The present study assesses the in vitro and in vivo bioavailability of genistein derivatives, hydroxyalkyl- and glycosyl alkyl ethers (glycoconjugates). Studies were carried out using compounds that exhibit higher in vitro antiproliferative activity in comparison with the parent isoflavone. Based on in vitro experiments using the Parallel Artificial Membrane Permeability Assay (PAMPA) and the Caco-2 cell monolayer permeability model, we found that modification of the isoflavone structure by O-alkylation improved bioavailability in comparison to genistein. Additionally, the structure of the substituent and its position on genistein influenced the type of mechanism involved in the transport of compounds through biological membranes. The PAMPA assay showed that the structure of glycoconjugates had a significant influence on the passive transport of the genistein synthetic derivatives through a biological membrane. Preferentially the glycoconjugates containing O-glycosidic bond were transported and the transport rate decreased as the carbon linker increased. For glycoconjugates, determination of their transport and metabolism through the Caco-2 membrane was not possible due to interaction with the membrane surface, probably by the change of compound structure caused by contact with the cells or degradation in medium. The intestinal absorption and metabolism of genistein and three derivatives, Ram-3, Ram'-3 and Ram-C-4α (Fig. 1), were tested in vivo in rats. We found that in comparison to genistein, glycoconjugates were metabolized more slowly and to a lesser extent. As part of the in vivo research, we performed analysis of compound levels in plasma samples after enzymatic hydrolysis, but in the collected samples, analytes were not observed. We hypothesize that glycoconjugates compounds bind plasma proteins and were removed from the sample. In conclusion, we show that O-functionalization of the natural, biologically active isoflavone genistein can affect biological activity, bioavailability, and the rate of compound metabolism. The position of the substituent, the length of the linker and the structure of sugar moieties provides a tool for the optimization of the derivative's biological properties.


Anticarcinogenic Agents/pharmacokinetics , Genistein/pharmacokinetics , Neoplasms/drug therapy , Administration, Oral , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/chemistry , Biological Availability , Caco-2 Cells , Cell Membrane Permeability , Female , Genistein/administration & dosage , Genistein/analogs & derivatives , Genistein/chemistry , Humans , Intestinal Absorption , Models, Animal , Molecular Structure , Permeability , Rats , Structure-Activity Relationship
10.
J Pharm Biomed Anal ; 177: 112809, 2020 Jan 05.
Article En | MEDLINE | ID: mdl-31541942

A simple and specific, rapid resolution liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for determination of chlorogenic acid in human plasma using neochlorogenic acid as the internal standard. Plasma samples were precipitated with methanol and separated on a Zorbax C18 column (50 × 2.1 mm, i.d. 1.8 µm) at a flow rate of 0.4 mL/min using a gradient mobile phase of methanol-water containing 0.1% formic acid (v/v). The detection was performed on a triple quadrupole tandem mass spectrometer by multiple reaction monitoring in negative ESI mode. The method was fully validated over the concentration range of 10-2000 ng/mL. The indicators of inter- and intra-day precision (RSD%) were all within 10.7%, and the accuracy (RE%) was ranged from -3.0% to 10.6%. Moreover, we evaluated this bioanalytical method by re-analysis of incurred samples as an additional measure of assay reproducibility. This method was successfully applied to pharmacokinetic study of CGA in Chinese subjects with advanced solid tumor after intramuscular injection administration of Chlorogenic acid for injection (CAFI).


Anticarcinogenic Agents/blood , Chlorogenic Acid/analogs & derivatives , High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Quinic Acid/analogs & derivatives , Tandem Mass Spectrometry/methods , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacokinetics , Area Under Curve , China , Chlorogenic Acid/administration & dosage , Chlorogenic Acid/blood , Chlorogenic Acid/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Stability , Humans , Injections, Intramuscular , Neoplasms/blood , Quinic Acid/administration & dosage , Quinic Acid/blood , Quinic Acid/pharmacokinetics , Reproducibility of Results , Sensitivity and Specificity
11.
PLoS One ; 14(12): e0226639, 2019.
Article En | MEDLINE | ID: mdl-31881053

Raloxifene is commonly used for breast cancer protection. The low bioavailability of raloxifene (2%) is the result of its low solubility and intestinal glucuronidation. The nano-lipid carriers are characterized by small particle size, biocompatibility, and sustainable properties that improve cellular uptake of the loaded drug. The aim of this study was the improvement of raloxifene bioavailability by enhancing its solubility and cellular penetration through formulation of D-α-tocopheryl polyethylene glycol 1000 succinate based transferosomes and augmenting their effect with the cationic cell-penetrating peptide transactivator of transcription of the human immunodeficiency virus. Particle size, zeta potential, and transmission electron microscope investigation of the formed nanocarriers were carried out. Ex vivo raloxifene permeation through rat skin and cell viability studies was investigated. The results of D-α-tocopheryl polyethylene glycol 1000 succinate- transactivator of transcription of the human immunodeficiency virus transferosomes showed an average vesicle size of 96.05 nm with positively charged vesicles 39.4 mV of zeta potential value. The results revealed significant (p < 0.05) enhancement of raloxifene permeation from raloxifene transferosomes- loaded film when compared with raw raloxifene film. IC50 results showed significant improvement of formulated raloxifene cytotoxicity by 1.42-fold in comparison with raw raloxifene against MCF-7 cell lines. The developed raloxifene-transferosomes are considered promising nano-lipid carriers for the enhancement delivery of raloxifene.


Drug Carriers/metabolism , Raloxifene Hydrochloride/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Vitamin E/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Administration, Cutaneous , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacokinetics , Anticarcinogenic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/prevention & control , Female , Humans , MCF-7 Cells , Raloxifene Hydrochloride/pharmacokinetics , Raloxifene Hydrochloride/pharmacology , Rats, Wistar , Selective Estrogen Receptor Modulators/pharmacokinetics , Selective Estrogen Receptor Modulators/pharmacology , Skin Absorption
12.
Biopharm Drug Dispos ; 40(5-6): 188-194, 2019 May.
Article En | MEDLINE | ID: mdl-31016737

Two indole compounds, indole-3-carbinol (I3C) and its acid condensation product, 3,3'-diindolymethane (DIM), have been shown to suppress the expression of flavin-containing monooxygenases (FMO) and to induce some hepatic cytochrome P450s (CYPs) in rats. In liver microsomes prepared from rats fed I3C or DIM, FMO-mediated nicotine N-oxygenation was decreased, whereas CYP-mediated nicotine metabolism to nicotine iminium and subsequently to cotinine was unchanged. Therefore, it was hypothesized that in mice DIM would also suppress nicotine N-oxygenation without affecting CYP-mediated nicotine metabolism. Liver microsomes were produced from male and female C57BL/6 J and CD1 mice fed 2500 parts per million (ppm) DIM for 14 days. In liver microsomes from DIM-fed mice, FMO-mediated nicotine N-oxygenation did not differ from the controls, but CYP-mediated nicotine metabolism was significantly increased, with results varying by sex and strain. To confirm the effects of DIM in vivo, control and DIM-fed CD1 male mice were injected subcutaneously with nicotine, and the plasma concentrations of nicotine, cotinine and nicotine-N-oxide were measured over 30 minutes. The DIM-fed mice showed greater cotinine concentrations compared with the controls 10 minutes following injection. It is concluded that the effects of DIM on nicotine metabolism in vitro and in vivo differ between mice and rats and between mouse strains, and that DIM is an effective inducer of CYP-mediated nicotine metabolism in commonly studied mouse strains.


Anticarcinogenic Agents/pharmacology , Indoles/pharmacology , Nicotine/pharmacokinetics , Animals , Animals, Outbred Strains , Anticarcinogenic Agents/pharmacokinetics , Brain/metabolism , Cotinine/blood , Female , Indoles/blood , Indoles/pharmacokinetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Nicotine/analogs & derivatives , Species Specificity
13.
Food Funct ; 10(2): 893-902, 2019 Feb 20.
Article En | MEDLINE | ID: mdl-30694275

Isothiocyanates from cruciferous vegetables are known for their potential anti-carcinogenic activities. These isothiocyanates are frequently consumed together as part of a regular diet, but their combined effects on carcinogenesis have not been well studied. Herein, we tested the hypothesis that combination of two isothiocyanates, i.e. allyl isothiocyanate and sulforaphane, produced a synergy in inhibiting the growth of A549 lung cancer cells. Our results showed that the combination treatment led to a stronger growth inhibition than the singular treatment. Isobologram analysis proved that the enhanced inhibitory effect of the combination treatment was synergistic. Flow cytometry demonstrated that the combination treatment caused more extensive cell cycle arrest and apoptosis than the singular treatment with modified expression of key proteins regulating these cellular processes. The combined treatment resulted in the production of intracellular reactive oxygen species, which might contribute to the inhibitory effects on cancer cells. Moreover, a synergy between allyl isothiocyanate and sulforaphane was also observed in anti-cell migration. Collectively, our results have demonstrated the potential of different isothiocyanates used in combination to produce enhanced protective effects against carcinogenesis.


Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Isothiocyanates/therapeutic use , A549 Cells , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Movement/drug effects , Drug Synergism , Food Preservatives/administration & dosage , Food Preservatives/pharmacokinetics , Food Preservatives/therapeutic use , Humans , Isothiocyanates/administration & dosage , Isothiocyanates/pharmacokinetics , Sulfoxides
14.
J Drug Target ; 27(4): 394-407, 2019 04.
Article En | MEDLINE | ID: mdl-30124078

Diets with naturally occuring chemopreventive agents are showing good potentials in serving dual purposes: firstly, for maintaining health, and secondly, for emerging as most puissant cost-effective strategy against chronic diseases like cancer. Genistein, one of the active soy isoflavone, is gaining attention due to its ability to impede carcinogenic processes by regulating wide range of associated molecules and signalling mechanisms. Epidemiologic and preclinical evidences suggest that sufficient consumption of soy-based food having genistein can be correlated to the reduction of cancer risk. However, certain adverse effects like poor oral bioavailability, low aqueous solubility and inefficient pharmacokinetics have pushed it down in the list of phytoconstituents currently undergoing successful clinical trials. In order to maximise the utilisation of therapeutic benefits of this phytoestrogen, suitable drug carrier designs are required. Recently, nanocarriers, mainly composed of polymeric materials, are progressively and innovatively exploited with the aim to improve pharmacokinetics and pharmacodynamics of genistein. Here, we have briefly reviewed (a) the targeted molecular mechanisms of geinstein, (b) nanopolymeric approaches opted so far in designing carriers and (c) the reasons behind their restricted clinical applications. Finally, some mechanism-based approaches are proposed presenting genistein as the future paradigm in cancer therapy.


Anticarcinogenic Agents/administration & dosage , Genistein/administration & dosage , Neoplasms/drug therapy , Animals , Anticarcinogenic Agents/pharmacokinetics , Biological Availability , Drug Carriers/chemistry , Drug Delivery Systems , Drug Design , Genistein/pharmacokinetics , Humans , Nanoparticles , Solubility
15.
Int J Mol Sci ; 19(9)2018 Sep 18.
Article En | MEDLINE | ID: mdl-30231546

Resveratrol is a naturally occurring polyphenol that provides several health benefits including cardioprotection and cancer prevention. However, its biological activity is limited by a poor bioavailability when taken orally. The aim of this work was to evaluate the capability of casein nanoparticles as oral carriers for resveratrol. Nanoparticles were prepared by a coacervation process, purified and dried by spray-drying. The mean size of nanoparticles was around 200 nm with a resveratrol payload close to 30 µg/mg nanoparticle. In vitro studies demonstrated that the resveratrol release from casein nanoparticles was not affected by the pH conditions and followed a zero-order kinetic. When nanoparticles were administered orally to rats, they remained within the gut, displaying an important capability to reach the intestinal epithelium. No evidence of nanoparticle "translocation" were observed. The resveratrol plasma levels were high and sustained for at least 8 h with a similar profile to that observed for the presence of the major metabolite in plasma. The oral bioavailability of resveratrol when loaded in casein nanoparticles was calculated to be 26.5%, 10 times higher than when the polyphenol was administered as oral solution. Finally, a good correlation between in vitro and in vivo data was observed.


Anticarcinogenic Agents/administration & dosage , Cardiotonic Agents/administration & dosage , Caseins/chemistry , Drug Carriers/chemistry , Nanoparticles/chemistry , Resveratrol/administration & dosage , Administration, Oral , Animals , Anticarcinogenic Agents/pharmacokinetics , Biological Availability , Cardiotonic Agents/pharmacokinetics , Male , Nanoparticles/ultrastructure , Rats, Wistar , Resveratrol/pharmacokinetics
16.
Life Sci ; 207: 340-349, 2018 Aug 15.
Article En | MEDLINE | ID: mdl-29959028

Cancer is one of the leading causes of death worldwide. Chemotherapy and radiotherapy are the conventional primary treatments for cancer patients. However, most of cancer cells develop resistance to both chemotherapy and radiotherapy after a period of treatment, besides their lethal side-effects. This motivated investigators to seek more effective alternatives with fewer side-effects. In the last few years, resveratrol, a natural polyphenolic phytoalexin, has attracted much attention due to its wide biological effects. In this concise review, we highlight the role of resveratrol in the prevention and therapy of cancer with particular focus on colorectal and skin cancer. Also, we discuss the molecular mechanisms underlying its chemopreventive and therapeutic activity. Finally, we highlight the problems associated with the clinical application of resveratrol and how attempts have been made to overcome these drawbacks.


Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/prevention & control , Skin Neoplasms/drug therapy , Skin Neoplasms/prevention & control , Stilbenes/therapeutic use , Animals , Anticarcinogenic Agents/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Autophagy , Cyclooxygenase Inhibitors/pharmacokinetics , Cyclooxygenase Inhibitors/therapeutic use , Female , Humans , Inflammasomes , Male , Mice , NF-E2-Related Factor 2/metabolism , Neoplasm Metastasis , Rats , Resveratrol , Sesquiterpenes , Stilbenes/pharmacokinetics , Phytoalexins
17.
World J Gastroenterol ; 24(16): 1679-1707, 2018 Apr 28.
Article En | MEDLINE | ID: mdl-29713125

Liver diseases are caused by different etiological agents, mainly alcohol consumption, viruses, drug intoxication or malnutrition. Frequently, liver diseases are initiated by oxidative stress and inflammation that lead to the excessive production of extracellular matrix (ECM), followed by a progression to fibrosis, cirrhosis and hepatocellular carcinoma (HCC). It has been reported that some natural products display hepatoprotective properties. Naringenin is a flavonoid with antioxidant, antifibrogenic, anti-inflammatory and anticancer properties that is capable of preventing liver damage caused by different agents. The main protective effects of naringenin in liver diseases are the inhibition of oxidative stress, transforming growth factor (TGF-ß) pathway and the prevention of the transdifferentiation of hepatic stellate cells (HSC), leading to decreased collagen synthesis. Other effects include the inhibition of the mitogen activated protein kinase (MAPK), toll-like receptor (TLR) and TGF-ß non-canonical pathways, the inhibition of which further results in a strong reduction in ECM synthesis and deposition. In addition, naringenin has shown beneficial effects on nonalcoholic fatty liver disease (NAFLD) through the regulation of lipid metabolism, modulating the synthesis and oxidation of lipids and cholesterol. Moreover, naringenin protects from HCC, since it inhibits growth factors such as TGF-ß and vascular endothelial growth factor (VEGF), inducing apoptosis and regulating MAPK pathways. Naringenin is safe and acts by targeting multiple proteins. However, it possesses low bioavailability and high intestinal metabolism. In this regard, formulations, such as nanoparticles or liposomes, have been developed to improve naringenin bioavailability. We conclude that naringenin should be considered in the future as an important candidate in the treatment of different liver diseases.


Anti-Inflammatory Agents/therapeutic use , Anticarcinogenic Agents/therapeutic use , Antioxidants/therapeutic use , Flavanones/therapeutic use , Liver Diseases/drug therapy , Liver/drug effects , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Anticarcinogenic Agents/adverse effects , Anticarcinogenic Agents/pharmacokinetics , Antioxidants/adverse effects , Antioxidants/pharmacokinetics , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Flavanones/adverse effects , Flavanones/pharmacokinetics , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Inflammation Mediators/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases/diagnosis , Liver Diseases/etiology , Liver Diseases/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects
18.
Drug Deliv Transl Res ; 8(2): 329-341, 2018 04.
Article En | MEDLINE | ID: mdl-28417445

Curcumin has shown promising inhibitory activity against HER-2-positive tumor cells in vitro but suffers from poor oral bioavailability in vivo. Our lab has previously developed a polymeric microparticle formulation for sustained delivery of curcumin for chemoprevention. The goal of this study was to examine the anticancer efficacy of curcumin-loaded polymeric microparticles in a transgenic mouse model of HER-2 cancer, Balb-neuT. Microparticles were injected monthly, and mice were examined for tumor appearance and growth. Initiating curcumin microparticle treatment at 2 or 4 weeks of age delayed tumor appearance by 2-3 weeks compared to that in control mice that received empty microparticles. At 12 weeks, abnormal (lobular hyperplasia, carcinoma in situ, and invasive carcinoma) mammary tissue area was significantly decreased in curcumin microparticle-treated mice, as was CD-31 staining. Curcumin treatment decreased mammary VEGF levels significantly, which likely contributed to slower tumor formation. When compared to saline controls, however, blank microparticles accelerated tumorigenesis and curcumin treatment abrogated this effect, suggesting that PLGA microparticles enhance tumorigenesis in this model. PLGA microparticle administration was shown to be associated with higher plasma lactic acid levels and increased activation of NF-κΒ. The unexpected side effects of PLGA microparticles may be related to the high dose of the microparticles that was needed to achieve sustained curcumin levels in vivo. Approaches that can decrease the overall dose of curcumin (for example, by increasing its potency or reducing its clearance rate) may allow the development of sustained release curcumin dosage forms as a practical approach to cancer chemoprevention.


Anticarcinogenic Agents/administration & dosage , Breast Neoplasms/drug therapy , Curcumin/administration & dosage , Drug Carriers/administration & dosage , Lactic Acid/administration & dosage , Polyglycolic Acid/administration & dosage , Animals , Anticarcinogenic Agents/pharmacokinetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Curcumin/pharmacokinetics , Cytokines/blood , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Disease Models, Animal , Drug Carriers/pharmacokinetics , Female , Genes, erbB-2 , Lactic Acid/blood , Lactic Acid/pharmacokinetics , Mice, Inbred BALB C , Mice, Transgenic , NF-kappa B/metabolism , Neovascularization, Pathologic/drug therapy , Polyglycolic Acid/pharmacokinetics , Polylactic Acid-Polyglycolic Acid Copolymer , Vascular Endothelial Growth Factor A/metabolism
19.
Bioorg Chem ; 76: 370-379, 2018 02.
Article En | MEDLINE | ID: mdl-29241109

Naproxen (nap) is belonging to Non-steriodal anti-inflammatory drugs (NSAIDs) group of drugs that characterized by their free carboxylic group. The therapeutic activity of nap is usually accompanied by GI untoward side effects. Recently synthesized naproxen amides of some amino acid esters prodrugs to mask the free carboxylic group were reported. Those prodrugs showed a promising colorectal cancer chemopreventive activity. The current study aims to investigate the fate and hydrolysis of the prodrugs kinetically in different pH conditions, simulated gastric and intestinal fluids with pHs of 1.2, 5.5 and 7.4 in vitro at 37 °C. The effect of enzymes on the hydrolysis of prodrugs was also studied through incubation of these prodrugs at 37 °C in human plasma and rat liver homogenates. The pharmacokinetic parameters of selected prodrugs and the liberated nap were studied after oral and intraperitoneal administration in male wistar rats. The results showed the hydrolysis of naproxen amides of amino acid esters to nap through two steps first by degradation of the ester moiety to form the amide of nap with amino acid and the second was through the degradation of the amide link to liberate nap. The two reactions were followed and studied kinetically where K1 and K2 (rate constants of degradation) is reported. The hydrolysis of prodrugs was faster in liver homogenates than in plasma. The relative bioavailability of the liberated nap in vivo was higher in case of prodrug containing ethyl glycinate moiety than that occupied l-valine ethyl ester moiety. Each of nap. prodrugs containing ethyl glycinate and l-valine ethyl ester moieties appears promising in liberating nap, decreasing direct GI side effect and consequently their colorectal cancer chemopreventive activity.


Amides/pharmacokinetics , Amino Acids/pharmacokinetics , Anticarcinogenic Agents/pharmacokinetics , Naproxen/analogs & derivatives , Naproxen/pharmacokinetics , Prodrugs/pharmacokinetics , Administration, Oral , Amides/administration & dosage , Amides/blood , Amides/chemistry , Amino Acids/administration & dosage , Amino Acids/blood , Amino Acids/chemistry , Animals , Anticarcinogenic Agents/administration & dosage , Anticarcinogenic Agents/blood , Anticarcinogenic Agents/chemistry , Colorectal Neoplasms/drug therapy , Drug Stability , Esters/administration & dosage , Esters/blood , Esters/chemistry , Esters/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Hydrolysis , Injections, Intraperitoneal , Kinetics , Liver/metabolism , Male , Naproxen/administration & dosage , Naproxen/blood , Prodrugs/administration & dosage , Prodrugs/analysis , Prodrugs/chemistry , Rats, Wistar
20.
Curr Med Chem ; 25(37): 4918-4928, 2018.
Article En | MEDLINE | ID: mdl-28545378

Recently many studies showed anticancer activities of piperine, a pungent alkaloid found in black pepper and some other Piper species. We attempted to summarize acquired data that support anticancer potential of this natural agent. Piperine has been reported to possess effective chemopreventive activity. It has been studied to affect via several mechanisms of action, in brief enhancing antioxidant system, increasing level and activity of detoxifying enzymes and suppressing stem cell self-renewal. Moreover, piperine has been found to inhibit proliferation and survival of various cancerous cell lines via modulating cell cycle progression and exhibiting anti-apoptotic activity, respectively. This compound has been shown to modify activity of various enzymes and transcription factors to inhibit invasion, metastasis and angiogenesis. Interestingly, piperine has exhibited antimutagenic activity and also inhibited activity and expression of multidrug resistance transporters such as P-gp and MRP-1. Besides, about all reviewed studies have reported selective cytotoxic activity of piperine on cancerous cells in compared with normal cells. Altogether, the studies completely underline promising candidacy of piperine for further development. The collected preclinical data we provided in this article can be useful in the design of future researches especially clinical trials with piperine.


Alkaloids/therapeutic use , Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Benzodioxoles/therapeutic use , Piperidines/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Alkaloids/biosynthesis , Alkaloids/metabolism , Alkaloids/pharmacology , Animals , Anticarcinogenic Agents/pharmacokinetics , Antimutagenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antioxidants/pharmacology , Benzodioxoles/metabolism , Benzodioxoles/pharmacology , Biological Availability , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Evaluation, Preclinical , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Inactivation, Metabolic/drug effects , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/prevention & control , Neoplasms/blood supply , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Piper/metabolism , Piperidines/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/metabolism , Polyunsaturated Alkamides/pharmacology , Stem Cells/cytology , Stem Cells/drug effects
...