Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.562
Filter
1.
Clin Pharmacol Ther ; 115(6): 1282-1292, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38264789

ABSTRACT

The discovery of circadian clock genes greatly amplified the study of diurnal variations impacting cancer therapy, transforming it into a rapidly growing field of research. Especially, use of chronomodulated treatment with 5-fluorouracil (5-FU) has gained significance. Studies indicate high interindividual variability (IIV) in diurnal variations in dihydropyrimidine dehydrogenase (DPD) activity - a key enzyme for 5-FU metabolism. However, the influence of individual DPD chronotypes on chronomodulated therapy remains unclear and warrants further investigation. To optimize precision dosing of chronomodulated 5-FU, this study aims to: (i) build physiologically-based pharmacokinetic (PBPK) models for 5-FU, uracil, and their metabolites, (ii) assess the impact of diurnal variation on DPD activity, (iii) estimate individual DPD chronotypes, and (iv) personalize chronomodulated 5-FU infusion rates based on a patient's DPD chronotype. Whole-body PBPK models were developed with PK-Sim(R) and MoBi(R). Sinusoidal functions were used to incorporate variations in enzyme activity and chronomodulated infusion rates as well as to estimate individual DPD chronotypes from DPYD mRNA expression or DPD enzymatic activity. Four whole-body PBPK models for 5-FU, uracil, and their metabolites were established utilizing data from 41 5-FU and 10 publicly available uracil studies. IIV in DPD chronotypes was assessed and personalized chronomodulated administrations were developed to achieve (i) comparable 5-FU peak plasma concentrations, (ii) comparable 5-FU exposure, and (iii) constant 5-FU plasma levels via "noise cancellation" chronomodulated infusion. The developed PBPK models capture the extent of diurnal variations in DPD activity and can help investigate individualized chronomodulated 5-FU therapy through testing alternative personalized dosing strategies.


Subject(s)
Antimetabolites, Antineoplastic , Circadian Rhythm , Dihydrouracil Dehydrogenase (NADP) , Fluorouracil , Models, Biological , Neoplasms , Precision Medicine , Fluorouracil/pharmacokinetics , Fluorouracil/administration & dosage , Humans , Dihydrouracil Dehydrogenase (NADP)/metabolism , Dihydrouracil Dehydrogenase (NADP)/genetics , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/administration & dosage , Precision Medicine/methods , Neoplasms/drug therapy , Circadian Rhythm/physiology , Drug Chronotherapy , Male , Female , Computer Simulation , Middle Aged , Uracil/pharmacokinetics , Uracil/administration & dosage , Uracil/analogs & derivatives
2.
Xenobiotica ; 53(10-11): 613-620, 2023.
Article in English | MEDLINE | ID: mdl-37966716

ABSTRACT

Dose adjustment based on renal function is essential in S-1, which contains the 5­fluorouracil prodrug tegafur, and platinum-based agent oxaliplatin (SOX) combination chemotherapy for colorectal cancer in patients with chronic kidney disease. However, limited evidence on dose adjustment in acute kidney injury (AKI) and challenges in determining dosing strategies. This study investigated the pharmacokinetics of SOX chemotherapy and renal biomarkers in rats.AKI was prepared by renal ischaemia-reperfusion injury in 1,2-dimethylhydrazine-induced colorectal cancer model rats. Serum creatinine (sCr) levels were determined as a renal biomarker. After administration of S-1 (2 mg/kg tegafur) and oxaliplatin (5 mg/kg), drug concentrations of tegafur, 5-FU, and platinum were measured in the plasma and tumours.No alterations in the area under the plasma concentration-time curve (AUC0-24h) values of 5-fluorouracil were observed between control and AKI model rats. The tumour concentrations of 5-fluorouracil in the mild and severe AKI groups were significantly lower than control group. The AUC0-24h for platinum increased with AKI severity. Notably, population pharmacokinetic analysis identified sCr as a covariate in platinum distribution after SOX chemotherapy.To optimise dose adjustment of SOX chemotherapy in patients with AKI, sCr may be a key factor in determining the appropriate dose.


Subject(s)
Acute Kidney Injury , Colorectal Neoplasms , Humans , Rats , Animals , Oxaliplatin , Tegafur/toxicity , Tegafur/pharmacokinetics , Fluorouracil/therapeutic use , Fluorouracil/pharmacokinetics , Kidney/pathology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/drug therapy , Biomarkers , Colorectal Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
3.
Cancer Chemother Pharmacol ; 92(2): 135-139, 2023 08.
Article in English | MEDLINE | ID: mdl-37351624

ABSTRACT

BACKGROUND: Capecitabine is an oral chemotherapy prodrug of 5-fluorouracil (5-FU) with unpredictable toxicity, especially in older adults. The aim of this study was to evaluate the pharmacokinetics (PK) of capecitabine and its metabolites in younger adults (< 70 years) and older adults (≥ 70 years) receiving capecitabine for solid cancer. METHODS: Eligible participants receiving capecitabine had 2 venous samples collected on day 14 of cycle 1 and cycle 2 of their treatment. Capecitabine and metabolite concentrations were determined using liquid chromatography with tandem mass spectrometry. A Bayesian estimation approach was used to generate individual estimates of PK parameters for 5-FU. A linear mixed-effect analysis of variance (ANOVA) model was used to compare dose-normalised log-transformed PK parameters between age groups. Correlations were determined by linear regression and logistic regression analyses. RESULTS: Of the total 26 participants, 58% were male with a median age of 67 years (range, 37-85) with 54% aged < 70 years and 46% aged ≥ 70 years. Participants aged ≥ 70 years, compared to those aged < 70 years, had a greater 5-FU exposure based on area under the concentration-time curve (AUC) of 17% (90% CI 103-134%; 0.893 vs. 0.762 mg h/L) and 14% increase in maximal concentration, Cmax (90% CI 82.1-159%; 0.343 vs. 0.300 mg/L). The 5-FU Cmax was positively associated with time up and go (TUG) (Pearson's correlation 0.77, p = 0.01), but not other geriatric assessment domains or severe toxicity. CONCLUSION: 5-FU exposure was significantly increased in older adults compared to younger adults receiving equivalent doses of capecitabine, and is a possible cause for increased toxicity in older adults.


Subject(s)
Deoxycytidine , Neoplasms , Male , Humans , Aged , Adult , Middle Aged , Aged, 80 and over , Female , Capecitabine/adverse effects , Pilot Projects , Bayes Theorem , Area Under Curve , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy
4.
Bioanalysis ; 14(1): 29-45, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34786955

ABSTRACT

Aim: To develop a new sensitive RP-HPLC method for simultaneous estimation of 5-fluorouracil (5-FU) and sonidegib (SDG). Materials & methods: Analytical and bioanalytical methods for simultaneous quantification of 5-FU and SDG in bulk, nanoformulations and in rat plasma were developed and validated using a gradient elution technique. Results: Separation of the analytes was effected on a Luna® C18 LC column using a mobile mixture comprising acetonitrile and acidified water. 5-FU and SDG were extracted from plasma matrix using liquid-liquid extraction. The applicability of the method was verified through single-dose oral pharmacokinetic study in Wistar rats. Conclusion: The developed methods allow a specific, sensitive and steady analytical procedure for the simultaneous estimation of 5-FU and SDG in nanoformulations and biological matrix.


Subject(s)
Biphenyl Compounds/pharmacokinetics , Fluorouracil/pharmacokinetics , Neoplasms/drug therapy , Pyridines/pharmacokinetics , Animals , Chromatography, High Pressure Liquid/methods , Male , Rats , Rats, Wistar
5.
Drug Deliv ; 28(1): 1569-1584, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34291722

ABSTRACT

To prepare glutaraldehyde-based cross-linked medium molecular weight chitosan nanoparticles encapsulated with 5-Fluorouracil (5-FU), to overcome dosing frequency as well as reducing acute oral toxicity and poor bioavailability of the drug. Medium molecular weight chitosan nanoparticles (MMWCH-NPs) were prepared by reverse micelles method based on glutaraldehyde (GA) cross-linking and optimized by the process as well as formulation variables like a various drug to polymer ratio, cross-linker volumes, varying stirring speeds (rpm), different time of rotation/stirring, respectively and their effects on the mean particles size distribution and entrapment efficiency %EE and %LC of NPs. Characterization of formulations was done by FTIR studies, TEM, PXRD, TGA, Stability, and dissolution drug release studies were performed by dialysis bag technique at both pH (1.2 & 7.4) and acute oral toxicity studies in albino rabbits. The formulated nanoparticles showed a smooth morphology with smaller particle size distribution (230-550 nm), zeta potential (-15 to -18 mV) required to achieve enhanced permeation and retention effect (EPR), entrapment efficiency (%EE 12-59%). These NPs exhibited a controlled drug release profile with 84.36% of the drug over a period of 24 h. Drug release data were fitted to different kinetic models which predominantly followed Fickian diffusion mechanism (R2 = 0.972-0.976, N = 0.326-0.256). The optimized formulation (5-FU6) was observed under DSC/TGA, TEM. PXRD curves, FTIR, which confirmed thermal stability, structural integrity, amorphous state, compatibility between drug and polymer of optimized (5-FU6) as well as reduced acute oral toxicity in albino rabbits. Cross-linked medium molecular weight chitosan nanoparticles are nontoxic, well-tolerated therefore could be the future candidate for therapeutic effects as novel drug delivery carrier for anticancer drug(s).


Subject(s)
Antineoplastic Agents/administration & dosage , Chitosan/chemistry , Fluorouracil/administration & dosage , Nanoparticles/chemistry , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Chemistry, Pharmaceutical , Delayed-Action Preparations , Drug Carriers , Drug Liberation , Drug Stability , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Glutaral/chemistry , Molecular Weight , Mouth Diseases/chemically induced , Mouth Diseases/prevention & control , Particle Size , Rabbits
6.
Cancer Chemother Pharmacol ; 88(5): 837-844, 2021 11.
Article in English | MEDLINE | ID: mdl-34331561

ABSTRACT

PURPOSE: Fluoropyrimidines are one of the most used drug class to treat cancer patients, although they show high levels of associated toxicity. This study analyzed 33 polymorphisms in 17 pharmacogenes involved with the pharmacogenomics of fluoropyrimidines, in gastrointestinal cancer patients undergoing fluoropyrimidine-based treatment in the Brazilian Amazon. METHODS: The study population was composed of 216 patients, 92 of whom have an anatomopathological diagnosis of gastric cancer and 124 of colorectal cancer. The single nucleotide polymorphisms (SNP) were genotyped by allelic discrimination using the TaqMan OpenArray Genotyping technology, with a panel of 32 customized assays, run in a QuantStudio ™ 12K Flex Real-Time PCR System (Applied Biosystems, Life Technologies, Carlsbad USA). Ancestry analysis was performed using 61 autosomal ancestry informative markers (AIMs). RESULTS: The study population show mean values of 48.1% European, 31.1% Amerindian, and 20.8% African ancestries. A significant risk association for general and severe toxicity was found in the rs4451422 of FPGS (p = 0.001; OR 3.40; CI 95% 1.65-7.00 and p = 0.006; OR 4.63; CI 95% 1.56-13.72, respectively) and the rs9524885 of ABCC4 (p = 0.023; OR 2.74; CI 95% 1.14-6.65 and p = 0.024; OR 5.36; IC 95% 1.24-23.11, respectively) genes. The rs760370 in the SLC29A1 gene (p = 0.009; OR 6.71; CI 95% 1.16-8.21) and the rs1801133 in the MTHFR toxicity (p = 0.023; OR 3.09; CI 95% 1.16-8.21) gene also demonstrated to be significant, although only for severe toxicity. The results found in this study did not have statistics analysis correction. CONCLUSION: Four polymorphisms of the ABCC4, FPGS, SLC29A1, and MTHFR genes are likely to be potential predictive biomarkers for precision medicine in fluoropyrimidine-based treatments in the population of the Brazilian Amazon, which is constituted by a unique genetic background.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/genetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brazil , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Equilibrative Nucleoside Transporter 1/genetics , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Fluorouracil/pharmacology , Humans , Leucovorin/pharmacology , Male , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Middle Aged , Multidrug Resistance-Associated Proteins/genetics , Organoplatinum Compounds/pharmacology , Peptide Synthases/genetics , Pharmacogenomic Variants , Polymorphism, Single Nucleotide
7.
Molecules ; 26(14)2021 Jul 13.
Article in English | MEDLINE | ID: mdl-34299532

ABSTRACT

Pharmacokinetic (PK) studies improve the design of dosing regimens in preclinical and clinical settings. In complex diseases like cancer, single-agent approaches are often insufficient for an effective treatment, and drug combination therapies can be implemented. In this work, in silico PK models were developed based on in vitro assays results, with the goal of predicting the in vivo performance of drug combinations in the context of cancer therapy. Combinations of reference drugs for cancer treatment, gemcitabine and 5-fluorouracil (5-FU), and repurposed drugs itraconazole, verapamil or tacrine, were evaluated in vitro. Then, two-compartment PK models were developed based on the previous in vitro studies and on the PK profile reported in the literature for human patients. Considering the quantification parameter area under the dose-response-time curve (AUCeffect) for the combinations effect, itraconazole was the most effective in combination with either reference anticancer drugs. In addition, cell growth inhibition was itraconazole-dose dependent and an increase in effect was predicted if itraconazole administration was continued (24-h dosing interval). This work demonstrates that in silico methods and AUCeffect are powerful tools to study relationships between tissue drug concentration and the percentage of cell growth inhibition over time.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Fluorouracil/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Computer Simulation , Deoxycytidine/pharmacokinetics , Deoxycytidine/pharmacology , Drug Combinations , Fluorouracil/pharmacokinetics , Humans , Models, Biological , Neoplasms/drug therapy , Gemcitabine
8.
Cancer Chemother Pharmacol ; 88(4): 619-631, 2021 10.
Article in English | MEDLINE | ID: mdl-34132895

ABSTRACT

PURPOSE: The aim was to investigate the pharmacokinetics of preoperatively administered intraperitoneal (IP) 5-FU in patients with resectable pancreatic ductal adenocarcinoma (PDAC) by analyzing levels of 5-FU and target metabolites in peritoneal fluid, plasma, liver, lymph nodes, pancreatic tumour, and pancreatic tissue. These results were correlated to expression of genes encoding enzymes of the 5-FU pathway and cell membrane transporters of 5-FU and FdUMP. METHODS: Twenty-two patients with PDAC were treated with IP 5-FU before surgery. The postoperative treatment followed a routine clinical protocol. 5-FU and its metabolites were analyzed by LC-MS/MS. The expression of genes encoding enzymes and transporters in the 5-FU pathway was analyzed by qPCR. RESULTS: After IP treatment, 5-FU could be detected in plasma, lymph nodes, liver, pancreatic tumour, and pancreatic tissue. The highest 5-FU concentration was found in the liver, also expressing high levels of the 5-FU transporter OAT2. 5-FU was converted to active FdUMP in all tissues and the highest concentration was measured in lymph nodes, liver and pancreatic tumour (18.5, 6.1 and 6.7 pmol/g, respectively). There was a correlation between the FdUMP and dUr levels in lymph nodes (r = 0.70, p = 0.0076). In tumours, there was an association between OAT2 expression and FdUMP concentration. CONCLUSION: The study shows uptake of IP 5-FU and drug metabolism to active FdUMP in pancreatic tumour, liver, and lymph nodes. Extended studies are warranted to evaluate the IP route for 5-FU administration in PDAC patients.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Carcinoma, Pancreatic Ductal/therapy , Fluorouracil/administration & dosage , Pancreatic Neoplasms/therapy , Aged , Antimetabolites, Antineoplastic/pharmacokinetics , Carcinoma, Pancreatic Ductal/pathology , Chromatography, Liquid , Combined Modality Therapy , Female , Fluorouracil/pharmacokinetics , Humans , Injections, Intraperitoneal , Male , Middle Aged , Pancreatic Neoplasms/pathology , Preoperative Care/methods , Tandem Mass Spectrometry , Tissue Distribution
9.
Cancer Med ; 10(14): 4781-4789, 2021 07.
Article in English | MEDLINE | ID: mdl-34121365

ABSTRACT

BACKGROUND: A low skeletal muscle mass (SMM) has been associated with increased toxicity and shorter survival in cancer patients treated with capecitabine, an oral prodrug of 5-fluorouracil (5-FU). Capecitabine and its metabolites are highly water-soluble and, therefore, more likely to distribute to lean tissues. The pharmacokinetics (PK) in patients with a low SMM could be changed, for example, by reaching higher maximum plasma concentrations. In this study, we aimed to examine whether the association between a low SMM and increased toxicity and shorter survival could be explained by altered PK of capecitabine and its metabolites. METHODS: Previously, a population PK model of capecitabine and metabolites in patients with solid tumors was developed. In our analysis, we included patients from this previous analysis for which evaluable abdominal computed tomography (CT)-scans were available. SMM was measured on CT-scans, by single slice evaluation at the third lumbar vertebra, using the Slice-o-Matic software. The previously developed population PK model was extended with SMM as a covariate, to assess the association between SMM and capecitabine and metabolite PK. RESULTS: PK and SMM data were available from 151 cancer patients with solid tumors. From the included patients, 55% had a low SMM. No relevant relationships were found between SMM and the PK parameters of capecitabine and, the active and toxic metabolite, 5-FU. SMM only correlated with the PK of the, most hydrophilic, but inactive and non-toxic, metabolite α-fluoro-ß-alanine (FBAL). Patients with a low SMM had a smaller apparent volume of distribution and lower apparent clearance of FBAL. CONCLUSIONS: No alterations in PK of capecitabine and the active and toxic metabolite 5-FU were observed in patients with a low SMM. Therefore, the previously identified increased toxicity and shorter survival in patients with a low SMM, could not be explained by changes in pharmacokinetic characteristics of capecitabine and metabolites.


Subject(s)
Antimetabolites, Antineoplastic/pharmacokinetics , Capecitabine/pharmacokinetics , Muscle, Skeletal/metabolism , Neoplasms/metabolism , Adult , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Capecitabine/administration & dosage , Capecitabine/adverse effects , Female , Fluorouracil/pharmacokinetics , Humans , Male , Middle Aged , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/pathology , Neoplasms/drug therapy , Neoplasms/mortality , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacokinetics , Sex Factors , Tomography, X-Ray Computed , Treatment Outcome , beta-Alanine/analogs & derivatives , beta-Alanine/pharmacokinetics
10.
Arch Toxicol ; 95(8): 2691-2718, 2021 08.
Article in English | MEDLINE | ID: mdl-34151400

ABSTRACT

5-Fluorouracil (5-FU) is a widely used chemotherapeutical that induces acute toxicity in the small and large intestine of patients. Symptoms can be severe and lead to the interruption of cancer treatments. However, there is limited understanding of the molecular mechanisms underlying 5-FU-induced intestinal toxicity. In this study, well-established 3D organoid models of human colon and small intestine (SI) were used to characterize 5-FU transcriptomic and metabolomic responses. Clinically relevant 5-FU concentrations for in vitro testing in organoids were established using physiologically based pharmacokinetic simulation of dosing regimens recommended for cancer patients, resulting in exposures to 10, 100 and 1000 µM. After treatment, different measurements were performed: cell viability and apoptosis; image analysis of cell morphological changes; RNA sequencing; and metabolome analysis of supernatant from organoids cultures. Based on analysis of the differentially expressed genes, the most prominent molecular pathways affected by 5-FU included cell cycle, p53 signalling, mitochondrial ATP synthesis and apoptosis. Short time-series expression miner demonstrated tissue-specific mechanisms affected by 5-FU, namely biosynthesis and transport of small molecules, and mRNA translation for colon; cell signalling mediated by Rho GTPases and fork-head box transcription factors for SI. Metabolomic analysis showed that in addition to the effects on TCA cycle and oxidative stress in both organoids, tissue-specific metabolic alterations were also induced by 5-FU. Multi-omics integration identified transcription factor E2F1, a regulator of cell cycle and apoptosis, as the best key node across all samples. These results provide new insights into 5-FU toxicity mechanisms and underline the relevance of human organoid models in the safety assessment in drug development.


Subject(s)
Colon/drug effects , Fluorouracil/toxicity , Intestine, Small/drug effects , Models, Biological , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Antimetabolites, Antineoplastic/toxicity , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Survival/drug effects , Colon/pathology , Dose-Response Relationship, Drug , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Humans , Intestine, Small/pathology , Male , Metabolomics , Organoids/drug effects , Oxidative Stress/drug effects , Transcriptome
11.
Eur J Pharm Biopharm ; 163: 223-239, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33864904

ABSTRACT

In the present study, combinatorial nanostructured lipid carrier gel of 5-fluorouracil and resveratrol was formulated, optimized and characterized to enhance permeation in between epidermis and dermis layers of the skin to obtain a synergistic effect against skin cancer. After extensive trials, a newly modified emulsiosonication method was developed and additionally, for the first time, stability studies were done in the beginning to optimize formulation technique, which exhibited two major benefits simultaneously; first, it provided best-optimized technique for preparation of combinatorial lipid-nanosystem, and secondly, it also demonstrated a detailed report card of durability of formulations. In vitro release study showed a significantly improved, slow and prolonged release of drugs from the optimized lipid-nanosystem (***p < 0.05), which followed non-Fickian Higuchi kinetics. Besides, mechanism of skin permeation enhancement study, dermatokinetic assessment, and depth analysis of optimized formulation on skin exhibited improved permeation and well distribution of drugs up to the dermis layer of skin. Moreover, combinatorial linogel possessed significantly greater efficacy (**p < 0.01) on the A431 cell line, as compared to the conventional formulation. Thus, findings revealed that modified method of preparation for dual drug-loaded lipid-nanosystem lead to the production of a stable formulation that also improved the retention of both 5-fluorouracil and resveratrol in between the epidermis and dermis region of skin thereby helping in the management and treatment of skin cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Drug Carriers/chemistry , Fluorouracil/administration & dosage , Resveratrol/administration & dosage , Skin Neoplasms/drug therapy , Administration, Cutaneous , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Drug Combinations , Drug Compounding/methods , Drug Liberation , Excipients/chemistry , Fluorouracil/pharmacokinetics , Humans , Lipids/chemistry , Nanoparticles/chemistry , Particle Size , Rats , Resveratrol/pharmacokinetics , Skin/metabolism , Skin/pathology , Skin Absorption , Skin Neoplasms/pathology , Tissue Distribution
12.
Hum Cell ; 34(4): 1153-1162, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33721218

ABSTRACT

As one of the most prevalent and deadly cancers worldwide, esophageal squamous cell carcinoma (ESCC) can be directly exposed to endocrine-disrupting chemical (EDC). As a potential EDC, diethylhexyl phthalate (DEHP) can trigger the development of various cancers, while the potential effect of DEHP on the ESCC progression was not clear. Our present study revealed that DEHP can trigger the proliferation of ESCC cells and decrease the cisplatin (CDDP) and fluorouracil (5-FU) sensitivity. Mechanistical studies indicated that DEHP can decrease the transcription of PTEN, a well-characterized tumor suppressor, in ESCC cells. Over expression of PTEN can reverse DEHP-regulated ESCC cell proliferation and chemosensitivity. Further, DEHP can increase the expression of HES-1, which can bind with the promoter of PTEN to inhibit its transcription. Collectively, DEHP can increase proliferation while decrease chemosensitivity of ESCC cells via regulation of HES-1/PTEN axis. Further, daily expression of DEHP may be a potent risk factor for ESCC development.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cisplatin/pharmacology , Diethylhexyl Phthalate/adverse effects , Endocrine Disruptors/adverse effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Fluorouracil/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Genes, Tumor Suppressor , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Transcription, Genetic/drug effects , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm/drug effects , Gene Expression/drug effects , Humans , Protein Binding , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism , Transcription, Genetic/genetics
13.
Int J Biol Macromol ; 178: 464-476, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33662416

ABSTRACT

Polymer-clay nanocomposite hydrogel films (PCNCHFs) were prepared from caboxymethyl cellulose, polyvinylpyrrolidone, agar and nanosepiolite clay (0, 0.3, 0.5, 0.7, 0.9 and 1.5% reinforcement) by treating thermally in a simple, rapid, and inexpensive route. The PCNCHFs and its 5-fluorouracil (FU)-loaded composites (PCNCHFs@FU) were tested for FU release and characterized by FTIR, XRD, FE-SEM, EDX, DSC, and TGA analyses to investigate their structural, morphological, and thermal properties. The nanosepiolite-loaded polymer composites (PCNCHF1 to PCNCHF5) exhibited higher tensile strength than the pristine polymer hydrogel (PCNCHF0); consequently, the thermal properties (glass- and melting-transition) were improved. The PCNCHFs@FU demonstrated prolonged FU release at pH 7.4 for 32 h. The biocompatibility of PCNCHFs was tested against human skin fibroblast (CCDK) cells. The viability of cells exposed to all PCNCHFs was >95% after 72 h of culture. The live/dead assay show the proliferation of fibroblast cells, confirming the biocompatibility of the hydrogels. The pH-sensitive PCNCHFs@FU release could be suitable for drug release in cancer therapy, and the developed PCNCHFs may also be useful for tissue engineering, food packaging, and other biological applications.


Subject(s)
Carboxymethylcellulose Sodium/chemistry , Drug Carriers/chemistry , Fluorouracil , Hydrogels/chemistry , Magnesium Silicates/chemistry , Nanocomposites/chemistry , Fluorouracil/chemistry , Fluorouracil/pharmacokinetics
14.
Cancer Res ; 81(7): 1654-1666, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33648930

ABSTRACT

Overcoming drug resistance is one of the biggest challenges in cancer chemotherapy. In this study, we examine whether targeting the long noncoding RNA taurine upregulated gene 1 (TUG1) could be an effective therapeutic approach to overcome drug resistance in pancreatic ductal adenocarcinoma (PDAC). TUG1 was expressed at significantly higher levels across 197 PDAC tissues compared with normal pancreatic tissues. Overall survival of patients with PDAC who had undergone 5-FU-based chemotherapy was shorter in high TUG1 group than in low TUG1 group. Mechanistically, TUG1 antagonized miR-376b-3p and upregulated dihydropyrimidine dehydrogenase (DPD). TUG1 depletion induced susceptibility to 5-FU in BxPC-3 and PK-9 pancreatic cell lines. Consistently, the cellular concentration of 5-FU was significantly higher under TUG1-depleted conditions. In PDAC xenograft models, intravenous treatment with a cancer-specific drug delivery system (TUG1-DDS) and 5-FU significantly suppressed PDAC tumor growth compared with 5-FU treatment alone. This novel approach using TUG1-DDS in combination with 5-FU may serve as an effective therapeutic option to attenuate DPD activity and meet appropriate 5-FU dosage requirements in targeted PDAC cells, which can reduce the systemic adverse effects of chemotherapy. SIGNIFICANCE: Targeting TUG1 coupled with a cancer-specific drug delivery system effectively modulates 5-FU catabolism in TUG1-overexpressing PDAC cells, thus contributing to a new combinatorial strategy for cancer treatment. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/7/1654/F1.large.jpg.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Oligonucleotides, Antisense/administration & dosage , Pancreatic Neoplasms/drug therapy , RNA, Long Noncoding/antagonists & inhibitors , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cohort Studies , Drug Delivery Systems/methods , Drug Synergism , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inactivation, Metabolic/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy/methods , Organ Specificity/drug effects , Organ Specificity/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/chemistry , RNA, Long Noncoding/drug effects , RNA, Long Noncoding/genetics , Xenograft Model Antitumor Assays
15.
Eur J Drug Metab Pharmacokinet ; 46(2): 317-324, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33619631

ABSTRACT

BACKGROUND AND OBJECTIVES: Irinotecan (CPT-11) is metabolized to an active metabolite 7-ethyl-10-hydroxycamptothecin (SN-38) by carboxylesterase (CES). SN-38 is then converted to the inactive metabolite SN-38 glucuronide (SN-38G) by glucuronosyltransferase 1A1 (UGT1A1). Genetic polymorphisms in UGT1A1 have been associated with altered SN-38 pharmacokinetics, which increase the risk of toxicity in patients. CPT-11 is also converted to 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin (APC) and 7-ethyl-10-(4-amino-1-piperidino) carbonyloxycamptothecin (NPC) by cytochrome P450 3A (CYP3A), and this route also affects the plasma concentration of SN-38. We evaluated the activities of UGT1A1, CYP3A, and CES and the factors affecting the pharmacokinetics of plasma SN-38 in patients with UGT1A1 gene polymorphisms. METHODS: Three male patients aged 56, 65, and 49 years were recruited for the analysis. All patients had pancreatic cancer, received FOLFIRINOX, and had UGT1A1*6/*6 (patients 1 and 3) or *6/*28 (patient 2) genetic polymorphisms. The rate constants for evaluating the enzyme activity were determined from the measured plasma concentration of CPT-11 and its metabolites using a two-compartment model by WinNonlin. RESULTS: The area under the plasma concentration-time curve (AUC) of SN-38 was patient 1 > patient 2 > patient 3. The rate constants obtained from the model analysis indicated the respective enzyme activities of UGT1A1 (k57), CYP3A (k13 + k19), and CES (k15). The order of values for UGT1A1 activity was patient 2 > patient 3 > patient 1. Since UGT1A1 activity was low in patient 1 with a high AUC of SN-38, it can be said that the increase in plasma concentration was due to a decrease in UGT1A1 activity. Conversely, the order of values for CYP3A and CES activities was patient 3 > patient 1 > patient 2 and patient 2 > patient 1 > patient 3, respectively. Patient 3 had the lowest AUC of SN-38, caused by a lower level of CES activity and increased CYP3A activity. CONCLUSION: In this study, we indicated that the plasma AUC of SN-38 and AUC ratio of SN-38G/SN-38 may depend on changes in the activities of CYP3A, CES, and UGT1A1. Using pharmacokinetic analysis, it is possible to directly evaluate enzyme activity and consider what kind of enzyme variation causes the increase in the AUC of SN-38.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Glucuronosyltransferase/genetics , Irinotecan/pharmacokinetics , Pancreatic Neoplasms/drug therapy , Aged , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Area Under Curve , Camptothecin/analogs & derivatives , Camptothecin/pharmacokinetics , Carboxylesterase/metabolism , Cytochrome P-450 CYP3A/metabolism , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Glucuronides/pharmacokinetics , Glucuronosyltransferase/metabolism , Humans , Irinotecan/administration & dosage , Leucovorin/administration & dosage , Leucovorin/pharmacokinetics , Male , Middle Aged , Models, Biological , Oxaliplatin/administration & dosage , Oxaliplatin/pharmacokinetics , Polymorphism, Genetic
16.
Clin Transl Sci ; 14(4): 1338-1348, 2021 07.
Article in English | MEDLINE | ID: mdl-33620159

ABSTRACT

Consensus guidelines exist for genotype-guided fluoropyrimidine dosing based on variation in the gene dihydropyrimidine dehydrogenase (DPYD). However, these guidelines have not been widely implemented in North America and most studies of pretreatment DPYD screening have been conducted in Europe. Given regional differences in treatment practices and rates of adverse events (AEs), we investigated the impact of pretreatment DPYD genotyping on AEs in a Canadian context. Patients referred for DPYD genotyping prior to fluoropyrimidine treatment were enrolled from December 2013 through November 2019 and followed until completion of fluoropyrimidine treatment. Patients were genotyped for DPYD c.1905+1G>A, c.2846A>T, c.1679T>G, and c.1236G>A. Genotype-guided dosing recommendations were informed by Clinical Pharmacogenetics Implementation Consortium guidelines. The primary outcome was the proportion of patients who experienced a severe fluoropyrimidine-related AE (grade ≥3, Common Terminology Criteria for Adverse Events version 5.0). Secondary outcomes included early severe AEs, severe AEs by toxicity category, discontinuation of fluoropyrimidine treatment due to AEs, and fluoropyrimidine-related death. Among 1394 patients, mean (SD) age was 64 (12) years, 764 (54.8%) were men, and 47 (3.4%) were DPYD variant carriers treated with dose reduction. Eleven variant carriers (23%) and 418 (31.0%) noncarriers experienced a severe fluoropyrimidine-related AE (p = 0.265). Six carriers (15%) and 284 noncarriers (21.1%) experienced early severe fluoropyrimidine-related AEs (p = 0.167). DPYD variant carriers treated with genotype-guided dosing did not experience an increased risk for severe AEs. Our data support a role for DPYD genotyping in the use of fluoropyrimidines in North America.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Dihydropyrimidine Dehydrogenase Deficiency/diagnosis , Dihydrouracil Dehydrogenase (NADP)/genetics , Neoplasms/drug therapy , Aged , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/pharmacokinetics , Canada , Capecitabine/administration & dosage , Capecitabine/adverse effects , Capecitabine/pharmacokinetics , Dihydropyrimidine Dehydrogenase Deficiency/genetics , Dihydrouracil Dehydrogenase (NADP)/metabolism , Female , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Fluorouracil/pharmacokinetics , Heterozygote , Humans , Male , Medical Oncology/standards , Middle Aged , Neoplasms/genetics , Pharmacogenomic Testing/standards , Pharmacogenomic Variants , Practice Guidelines as Topic , Precision Medicine/standards , Precision Medicine/statistics & numerical data , Retrospective Studies
17.
J Chemother ; 33(1): 51-55, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32066346

ABSTRACT

We administered FOLFOX (oxaliplatin (L-OHP) plus infusional 5-fluorouracil (5-FU) and leucovorin) to an hemodialysis (HD) patient with advanced gastric cancer (AGC), and investigated pharmacokinetics (PKs) and dialyzability of L-OHP. The patient was a 54-year-old Japanese man with a diagnosis of inoperable AGC. FOLFOX was instituted 3 h prior to the start of a 4 h HD period with the L-OHP and 5-FU doses reduced by 50% for the first cycle, and 30% reduced dose was administered for the second cycle. We performed an analysis of the PKs of L-OHP during these two cycles. Volume of distribution and area under the curve of the 30% reduced L-OHP dose were 56.7 L and 30.0 µg·h/mL, respectively. A dose reduction of L-OHP by 30%-50% may be advisable for the initial administration, given the need for careful administration of chemotherapy in HD patients, with particular attention to the development of hematological toxicities and neuropathy.


Subject(s)
Kidney Failure, Chronic/therapy , Oxaliplatin/pharmacokinetics , Oxaliplatin/therapeutic use , Renal Dialysis , Stomach Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dose-Response Relationship, Drug , Fluorouracil/pharmacokinetics , Fluorouracil/therapeutic use , Humans , Kidney Failure, Chronic/metabolism , Leucovorin/pharmacokinetics , Leucovorin/therapeutic use , Male , Middle Aged , Organoplatinum Compounds/pharmacokinetics , Organoplatinum Compounds/therapeutic use
18.
Eur J Pharm Biopharm ; 158: 371-378, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33309846

ABSTRACT

The encapsulation of nanoparticles within microparticles designed for specific delivery to the colon is a relevant strategy to avoid premature degradation or release of nanoparticles during their passage through the stomach and upper gastrointestinal tract (GIT), allowing the targeted delivery of chemotherapeutics to the colon after oral administration. Here, we designed an oral multiparticulate system to achieve targeted release in the colon. In this sense, chitosan nanoparticles (CS NPs) encapsulated with 5-fluorouracil (5-FU) and incorporated into retrograded starch and pectin (RS/P) microparticles were developed and their in vivo distribution along the mouse GIT after oral administration was monitored using multispectral optical imaging. In vitro release studies revealed that the encapsulation of CS NPs into RS/P microparticles promoted greater control of 5-FU release rates, with a significant reduction (53%) in acid media that might replicate that found in the stomach following oral administration. The evaluation of the in vivo biodistribution of the CS NPs in mice showed a faster clearance in the distribution pattern along the mouse GIT, i.e., a shorter transit time of CS NPs compared to CS NPs-loaded RS/P microparticles. Additionally, CS NPs alone showed non-specific absorption into the blood-stream with associated kidney accumulation, while for the CS NPs-loaded RS/P microparticles no significant accumulation was observed in blood or major clearance organs. This suggests the specific degradability of RS/P by the colon microbiota appears to have been decisive in the higher protection of the CS NPs along the GIT until release in the colon, preventing unwanted absorption into the bloodstream and major organs following oral administration. Our findings represent a proof of concept for the use of RS/P microparticles as potential carriers for delivering drug-loaded nanoparticles to the colon and this work will contribute to the development of oral-systems for colorectal cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Colorectal Neoplasms/drug therapy , Drug Carriers/chemistry , Drug Compounding/methods , Nanoparticles/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Biological Availability , Chitosan/administration & dosage , Colon/metabolism , Colon/microbiology , Drug Carriers/metabolism , Drug Liberation , Female , Fluorouracil/administration & dosage , Fluorouracil/pharmacokinetics , Gastrointestinal Microbiome/physiology , Humans , Intestinal Absorption , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Models, Animal , Particle Size , Pectins/chemistry , Pectins/metabolism , Proof of Concept Study , Starch/chemistry , Starch/metabolism , Tissue Distribution
20.
Anticancer Res ; 40(12): 6585-6597, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33288553

ABSTRACT

Recently, therapeutic drug monitoring of 5-fluorouracil (5-FU), the key chemotherapeutic drug for colorectal cancer, has been applied in daily clinical practice and has contributed towards improving clinical outcomes. However, current dose modifications are based only on values of the area under the plasma concentration-time profile, which are simply calculated from plasma 5-FU concentrations and infusion periods. When dose-limiting toxicities occur, the dosing is empirically reduced or discontinued, leading to treatment failure. To prevent this predictable failure and obtain better clinical outcomes, rational dosage-based strategies are required for 5-FU. Combining therapeutic drug monitoring with a mathematical approach using a pharmacokinetic- pharmacodynamic/toxicodynamic model is expected to help simulate time-course profiles of the efficacy of drugs and the degree of toxicity, thereby contributing towards dose setting for individual patients. Therefore, to facilitate pharmacometric modelling and simulation techniques for optimising current oncology therapies, this review focuses on pharmacometrics approaches for personalizing 5-FU-based chemotherapy.


Subject(s)
Fluorouracil/pharmacology , Fluorouracil/pharmacokinetics , Precision Medicine , Circadian Rhythm , Fluorouracil/blood , Fluorouracil/toxicity , Humans , Models, Biological , Prodrugs/pharmacokinetics , Prodrugs/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...