Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Biomater Adv ; 155: 213660, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37976832

ABSTRACT

A major obstacle for chemotherapeutics in Glioblastoma (GB) is to reach the tumour cells due to the presence of the blood-brain barrier (BBB) and chemoresistance of anticancer drugs. The present study reports two polyunsaturated fatty acids, gamma-linolenic acid (GLA) and alpha-linolenic acid (ALA) appended nanostructured lipid carriers (NLCs) of a CNS negative chemotherapeutic drug docetaxel (DTX) for targeted delivery to GB. The ligand appended DTX-NLCs demonstrated particle size < 160 nm, PDI < 0.29 and a negative surface charge. The successful linkage of GLA (41 %) and ALA (30 %) ligand conjugation to DTX- NLCs was confirmed by diminished surface amino groups on the NLCs, lower surface charge and FTIR profiling. Fluorophore labelled GLA-DTX-NLCs and ALA-DTX-NLCs permeated the in-vitro 3D BBB model with Papp values of 1.8 × 10-3 and 1.9 × 10-3 cm/s respectively. Following permeation, both formulations showed enhanced uptake by GB immortalised cells while ALA-DTX-NLCs showed higher uptake in patient-derived GB cells as evidenced in an in-vitro 3D blood brain tumour barrier (BBTB) model. Both surface functionalised formulations showed higher internalisation in GB cells as compared to bare DTX-NLCs. ALA-DTX-NLCs and GLA-DTX-NLCs showed 13.9-fold and 6.8-fold higher DTX activity respectively at 24 h as indicated by IC50 values when tested in patient-derived GB cells. ALA-DTX-NLCs displayed better efficacy than GLA-DTX-NLCs when tested against 3D tumour spheroids and patient-derived cells. These novel formulations will contribute widely to overcoming biological barriers for treating glioblastoma.


Subject(s)
Drug Carriers , Glioblastoma , Humans , Glioblastoma/drug therapy , Blood-Brain Barrier , Ligands , Lipids/therapeutic use , Docetaxel , Fatty Acids, Unsaturated/therapeutic use
2.
Spectrochim Acta A Mol Biomol Spectrosc ; 273: 121018, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35189493

ABSTRACT

Meningiomas remains a clinical dilemma. They are the commonest "benign" types of brain tumours and, although being typically benign, they are divided into three WHO grades categories (I, II and III) which are associated with the tumour growth rate and likelihood of recurrence. Recurrence depends on extend of surgery as well as histopathological diagnosis. There is a marked variation amongst surgeons in the follow-up arrangements for their patients even within the same unit which has a significant clinical, and financial implication. Knowing the tumour grade rapidly is an important factor to predict surgical outcomes and adequate patient treatment. Clinical follow up sometimes is haphazard and not based on clear evidence. Spectrochemical techniques are a powerful tool for cancer diagnostics. Raman hyperspectral imaging is able to generate spatially-distributed spectrochemical signatures with great sensitivity. Using this technique, 95 brain tissue samples (66 meningiomas WHO grade I, 24 meningiomas WHO grade II and 5 meningiomas that reoccurred) were analysed in order to discriminate grade I and grade II samples. Newly-developed three-dimensional discriminant analysis algorithms were used to process the hyperspectral imaging data in a 3D fashion. Three-dimensional principal component analysis quadratic discriminant analysis (3D-PCA-QDA) was able to distinguish grade I and grade II meningioma samples with 96% test accuracy (100% sensitivity and 95% specificity). This technique is here shown to be a high-throughput, reagent-free, non-destructive, and can give accurate predictive information regarding the meningioma tumour grade, hence, having enormous clinical potential with regards to being developed for intra-operative real-time assessment of disease.


Subject(s)
Brain Neoplasms , Meningeal Neoplasms , Meningioma , Brain Neoplasms/diagnostic imaging , Child , Discriminant Analysis , Humans , Hyperspectral Imaging , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/pathology , Meningioma/diagnostic imaging , Meningioma/pathology
3.
J Alzheimers Dis ; 86(1): 343-364, 2022.
Article in English | MEDLINE | ID: mdl-35034897

ABSTRACT

BACKGROUND: The effects of the key pathogens and virulence factors associated with gum disease such as Porphyromonas gingivalis (P. gingivalis) on the central nervous system is of great interest with respect to development of neuropathologies and hence therapeutics and preventative strategies. Chronic infections and associated inflammation are known to weaken the first line of defense for the brain, the blood-brain barrier (BBB). OBJECTIVE: The focus of this study is to utilize an established human in vitro BBB model to evaluate the effects of P. gingivalis virulence factors lipopolysaccharide (LPS) and outer membrane vesicles (OMVs) on a primary-derived human model representing the neurovascular unit of the BBB. METHODS: Changes to the integrity of the BBB after application of P. gingivalis LPS and OMVs were investigated and correlated with transport of LPS. Additionally, the effect of P. gingivalis LPS and OMVs on human brain microvascular endothelial cells in monolayer was evaluated using immunofluorescence microscopy. RESULTS: The integrity of the BBB model was weakened by application of P. gingivalis LPS and OMVs, as measured by a decrease in electrical resistance and a recovery deficit was seen in comparison to the controls. Application of P. gingivalis OMVs to a monoculture of human brain microvascular endothelial cells showed disruption of the tight junction zona occludens protein (ZO-1) compared to controls. CONCLUSION: These findings show that the integrity of tight junctions of the human BBB could be weakened by association with P. gingivalis virulence factors LPS and OMVs containing proteolytic enzymes (gingipains).


Subject(s)
Lipopolysaccharides , Porphyromonas gingivalis , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Humans , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Permeability , Tight Junction Proteins/metabolism , Virulence Factors
4.
J Alzheimers Dis ; 85(3): 957-973, 2022.
Article in English | MEDLINE | ID: mdl-34897087

ABSTRACT

The central nervous system (CNS) is protected by a highly selective barrier, the blood-brain barrier (BBB), that regulates the exchange and homeostasis of bloodborne molecules, excluding xenobiotics. This barrier forms the first line of defense by prohibiting pathogens from crossing to the CNS. Aging and chronic exposure of the BBB to pathogens renders it permeable, and this may give rise to pathology in the CNS such as Alzheimer's disease (AD). Researchers have linked pathogens associated with periodontitis to neuroinflammation and AD-like pathology in vivo and in vitro. Although the presence of periodontitis-associated bacteria has been linked to AD in several clinical studies as DNA and virulence factors were confirmed in brain samples of human AD subjects, the mechanism by which the bacteria traverse to the brain and potentially influences neuropathology is unknown. In this review, we present current knowledge about the association between periodontitis and AD, the mechanism whereby periodontal pathogens might provoke neuroinflammation and how periodontal pathogens could affect the BBB. We suggest future studies, with emphasis on the use of human in vitro models of cells associated with the BBB to unravel the pathway of entry for these bacteria to the CNS and to reveal the molecular and cellular pathways involved in initiating the AD-like pathology. In conclusion, evidence demonstrates that bacteria associated with periodontitis and their virulence factors are capable of inflecting damage to the BBB and have a role in giving rise to pathology similar to that found in AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Blood-Brain Barrier/metabolism , Periodontitis , Virulence Factors , Alzheimer Disease/blood , Animals , Biological Transport , Brain/metabolism , Brain/pathology , Humans , Periodontitis/complications , Periodontitis/metabolism , Virulence Factors/blood
5.
Mater Sci Eng C Mater Biol Appl ; 121: 111774, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33579439

ABSTRACT

The blood-brain barrier (BBB) and blood-brain tumour barrier (BBTB) pose a significant challenge to drug delivery to brain tumours, including aggressive glioblastoma (GB). The present study rationally designed functional nanostructured lipid carriers (NLC) to tailor their BBB penetrating properties with high encapsulation of CNS negative chemotherapeutic drug docetaxel (DTX). We investigated the effect of four liquid lipids, propylene glycol monolaurate (Lauroglycol® 90), Capryol® propylene glycol monocaprylate, caprylocaproylmacrogol-8-glycerides (Labrasol®) and polyoxyl-15-hydroxystearate (Kolliphor® HS15) individually and in combination to develop NLCs with effective permeation across in-vitro 3D BBB model without alteration in the integrity of the barrier. With desirable spherical shape as revealed by TEM and an average particle size of 123.3 ± 0.642 nm and zeta potential of -32 mV, DTX-NLCs demonstrated excellent stability for six months in its freeze-dried form. The confocal microscopy along with flow cytometry data revealed higher internalisation of DTX-NLCs in U87MG over SVG P12 cells. Micropinocytosis was observed to be one of the dominant pathways for internalisation in U87MG cells while clathrin-mediated pathway was more predominat in patient-derived glioblastoma cells. The NLCs readily penetrated the actively proliferating peripheral cells on the surface of the 3D tumour spheroids as compared to the necrotic core. The DTX-NLCs induced cell arrest through G2/M phase with a significant decrease in the mitochondrial reserve capacity of cells. The NLCs circumvented BBTB with high permeability followed by accumulation in glioblastoma cells with patient-derived cells displaying ~2.4-fold higher uptake in comparison to U87MG when studied in a 3D in-vitro model of BBTB/GB. We envisage this simple and industrially feasible technology as a potential candidate to be developed as GB nanomedicine.


Subject(s)
Glioblastoma , Nanostructures , Blood-Brain Barrier , Drug Carriers/therapeutic use , Drug Delivery Systems , Glioblastoma/drug therapy , Humans , Lipids/therapeutic use , Particle Size , Permeability
6.
J Med Chem ; 64(3): 1497-1509, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33499592

ABSTRACT

For the first time, the in silico design, screening, and in vitro validation of potent GSK-3ß type-II inhibitors are presented. In the absence of crystallographic evidence for a DFG-out GSK-3ß activation loop conformation, computational models were designed using an adapted DOLPHIN approach and a method consisting of Prime loop refinement, induced-fit docking, and molecular dynamics. Virtual screening of the Biogenics subset from the ZINC database led to an initial selection of 20 Phase I compounds revealing two low micromolar inhibitors in an isolated enzyme assay. Twenty more analogues (Phase II compounds) related to the hit [pyrimidin-2-yl]amino-furo[3,2-b]furyl-urea scaffold were selected for structure-activity relationship analysis. The Phase II studies led to five highly potent nanomolar inhibitors, with compound 23 (IC50 =0.087 µM) > 100 times more potent than the best Phase I inhibitor, and selectivity for GSK-3ß inhibition compared to homologous kinases was observed. Ex vivo experiments (SH-SY5Y cell lines) for tau hyperphosphorylation revealed promising neuroprotective effects at low micromolar concentrations. The type-II inhibitor design has been unraveled as a potential route toward more clinically effective GSK-3ß inhibitors.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Cell Line, Tumor , Drug Design , Humans , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Phosphorylation , Structure-Activity Relationship , Substrate Specificity , tau Proteins/biosynthesis , tau Proteins/genetics
7.
J Control Release ; 238: 43-57, 2016 09 28.
Article in English | MEDLINE | ID: mdl-27448441

ABSTRACT

A minor population of glioblastoma stem-like cells (GSCs) has been implicated in the relapse and resistance of glioblastoma to therapeutic treatments. Based on knowledge of the involvement of multiple microRNAs in GSC propagation, we designed a combinational approach to target the GSC population with multiple miRNA-based therapeutics. As carriers for the targeted delivery we took advantage of two aptamers that bind to, and inhibit, the receptor tyrosine kinases, Axl and PDGFRß. We showed that the aptamer conjugates are transported through an in vitro blood-brain barrier (BBB) model. Furthermore, combining miR-137 and antimiR-10b synergizes with the receptor inhibitory function of aptamer carriers and prevents GSC expansion. Results highlighted the potential of combining multifunctional RNA-based therapeutics for selective targeting of GSCs and offer a proof of principle strategy to potentially fulfill the still unmet need for effective and safe treatment of glioma.


Subject(s)
Antagomirs/therapeutic use , Aptamers, Nucleotide/therapeutic use , Brain Neoplasms/therapy , Genetic Therapy/methods , Glioma/therapy , MicroRNAs/antagonists & inhibitors , MicroRNAs/therapeutic use , Neoplastic Stem Cells/pathology , Antagomirs/genetics , Aptamers, Nucleotide/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Transfer Techniques , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , MicroRNAs/genetics , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Receptor, Platelet-Derived Growth Factor beta/metabolism , Axl Receptor Tyrosine Kinase
8.
PLoS One ; 10(8): e0134957, 2015.
Article in English | MEDLINE | ID: mdl-26252900

ABSTRACT

Malignant glioma is characterised by a rapid growth rate and high capacity for invasive infiltration to surrounding brain tissue; hence, diagnosis and treatment is difficult and patient survival is poor. Aptamers contribute a promising and unique technology for the in vitro imaging of live cells and tissues, with a potentially bright future in clinical diagnostics and therapeutics for malignant glioma. The binding selectivity, uptake capacity and binding target of two DNA aptamers, SA43 and SA44, were investigated in glioma cells and patient tissues. The binding assay showed that SA43 and SA44 bound with strong affinity (Kd, 21.56 ± 4.60 nM and Kd, 21.11 ± 3.30 nM respectively) to the target U87MG cells. Quantitative analysis by flow cytometry showed that the aptamers were able to actively internalise in U87MG and 1321N1 glioma cells compared to the non-cancerous and non-glioma cell types. Confocal microscopy confirmed staining in the cytoplasm, and co-localisation studies with endoplasmic reticulum, Golgi apparatus and lysosomal markers suggested internalisation and compartmentalisation within the endomembrane system. Both aptamers selectively bound to Ku 70 and Ku 80 DNA repair proteins as determined by aptoprecipitation (AP) followed by mass spectrometry analysis and confirmation by Western blot. In addition, aptohistochemical (AHC) staining on paraffin embedded, formalin fixed patient tissues revealed that the binding selectivity was significantly higher for SA43 aptamer in glioma tissues (grade I, II, III and IV) compared to the non-cancerous tissues, whereas SA44 did not show selectivity towards glioma tissues. The results indicate that SA43 aptamer can differentiate between glioma and non-cancerous cells and tissues and therefore, shows promise for histological diagnosis of glioma.


Subject(s)
Aptamers, Nucleotide/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Aptamers, Nucleotide/chemistry , Biotinylation , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival , Chemical Precipitation , Flow Cytometry , Glioma/pathology , Humans , Immunohistochemistry , Microscopy, Confocal , Neoplasm Proteins/metabolism , Nucleic Acid Conformation , Subcellular Fractions/metabolism , Temperature
9.
Mol Oncol ; 9(3): 704-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25499534

ABSTRACT

BACKGROUND: Glioblastoma is the most aggressive primary brain tumor, and is associated with a very poor prognosis. In this study we investigated the potential of microRNA expression profiles to predict survival in this challenging disease. METHODS: MicroRNA and mRNA expression data from glioblastoma (n = 475) and grade II and III glioma (n = 178) were accessed from The Cancer Genome Atlas. LASSO regression models were used to identify a prognostic microRNA signature. Functionally relevant targets of microRNAs were determined using microRNA target prediction, experimental validation and correlation of microRNA and mRNA expression data. RESULTS: A 9-microRNA prognostic signature was identified which stratified patients into risk groups strongly associated with survival (p = 2.26e-09), significant in all glioblastoma subtypes except the non-G-CIMP proneural group. The statistical significance of the microRNA signature was higher than MGMT methylation in temozolomide treated tumors. The 9-microRNA risk score was validated in an independent dataset (p = 4.50e-02) and also stratified patients into high- and low-risk groups in lower grade glioma (p = 5.20e-03). The majority of the 9 microRNAs have been previously linked to glioblastoma biology or treatment response. Integration of the expression patterns of predicted microRNA targets revealed a number of relevant microRNA/target pairs, which were validated in cell lines. CONCLUSIONS: We have identified a novel, biologically relevant microRNA signature that stratifies high- and low-risk patients in glioblastoma. MicroRNA/mRNA interactions identified within the signature point to novel regulatory networks. This is the first study to formulate a survival risk score for glioblastoma which consists of microRNAs associated with glioblastoma biology and/or treatment response, indicating a functionally relevant signature.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , MicroRNAs/genetics , Aged , Brain Neoplasms/drug therapy , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/drug therapy , Humans , Male , MicroRNAs/metabolism , Middle Aged , Neoplasm Grading , Prognosis , Regression Analysis , Risk Factors , Survival Analysis , Temozolomide , Treatment Outcome
10.
Mol Neurobiol ; 50(2): 545-58, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24696266

ABSTRACT

The need for glioma biomarkers with improved sensitivity and specificity has sparked research into short non-coding RNA known as microRNA (miRNA). Altered miRNA biogenesis and expression in glioma plays a vital role in important signaling pathways associated with a range of tumor characteristics including gliomagenesis, invasion, and malignancy. This review will discuss current research into the role of miRNA in glioma and altered miRNA expression in biofluids as candidate biomarkers with a particular focus on glioblastoma, the most malignant form of glioma. The isolation and characterization of miRNA using cellular and molecular biology techniques from the circulation of glioma patients could potentially be used for improved diagnosis, prognosis, and treatment decisions. We aim to highlight the links between research into miRNA function, their use as biomarkers, and how these biomarkers can be used to predict response to therapy. Furthermore, increased understanding of miRNA in glioma biology through biomarker research has led to the development of miRNA therapeutics which could restore normal miRNA expression and function and improve the prognosis of glioma patients. A panel of important miRNA biomarkers for glioma in various biofluids discovered to date has been summarized here. There is still a need, however, to standardize techniques for biomarker characterization to bring us closer to clinically relevant miRNA-based diagnostic and therapeutic signatures. A clinically validated biomarker panel has potential to improve time to diagnosis, predicting response to treatment and ultimately the prognosis of glioma patients.


Subject(s)
Biomarkers/analysis , Brain Neoplasms/diagnosis , Glioma/diagnosis , MicroRNAs , Signal Transduction/genetics , Animals , Brain Neoplasms/blood , Brain Neoplasms/cerebrospinal fluid , Brain Neoplasms/therapy , Glioma/blood , Glioma/cerebrospinal fluid , Glioma/therapy , Humans , MicroRNAs/blood , MicroRNAs/cerebrospinal fluid , Prognosis , Signal Transduction/physiology
11.
Pharmacogenomics ; 12(6): 815-26, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21692613

ABSTRACT

AIM: To conduct a pragmatic, randomized controlled trial to assess whether thiopurine methyltransferase (TPMT) genotyping prior to azathioprine reduces adverse drug reactions (ADRs). METHODS: A total of 333 participants were randomized 1:1 to undergo TPMT genotyping prior to azathioprine or to commence treatment without genotyping. RESULTS: There was no difference in the primary outcome of stopping azathioprine due to an adverse reaction (ADR, p = 0.59) between the two study arms. ADRs were more common in older patients (p = 0.01). There was no increase in stopping azathioprine due to ADRs in TPMT heterozygotes compared with wild-type individuals. The single individual with TPMT variant homozygosity experienced severe neutropenia. CONCLUSION: Our work supports the strong evidence that individuals with TPMT variant homozygosity are at high risk of severe neutropenia, whereas TPMT heterozygotes are not at increased risk of ADRs at standard doses of azathioprine.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Azathioprine/administration & dosage , Azathioprine/adverse effects , Methyltransferases/genetics , Adult , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/prevention & control , Genetic Predisposition to Disease , Genetic Variation , Genotype , Heterozygote , Homozygote , Humans , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/genetics , Neutropenia/genetics , Phenotype
12.
Eur J Pharm Sci ; 43(3): 141-50, 2011 Jun 14.
Article in English | MEDLINE | ID: mdl-21540108

ABSTRACT

A clinical study was conducted to assess the ability of a microdose (100 µg) to predict the human pharmacokinetics (PK) following a therapeutic dose of clarithromycin, sumatriptan, propafenone, paracetamol (acetaminophen) and phenobarbital, both within the study and by reference to the existing literature on these compounds and to explore the source of any nonlinearity if seen. For each drug, 6 healthy male volunteers were dosed with 100 µg (14)C-labelled compound. For clarithromycin, sumatriptan, and propafenone this labelled dose was administered alone, i.e. as a microdose, orally and intravenously (iv) and as an iv tracer dose concomitantly with an oral non-labelled therapeutic dose, in a 3-way cross over design. The oral therapeutic doses were 250, 50, and 150 mg, respectively. Paracetamol was given as the labelled microdose orally and iv using a 2-way cross over design, whereas phenobarbital was given only as the microdose orally. Plasma concentrations of total (14)C and parent drug were measured using accelerator mass spectrometry (AMS) or HPLC followed by AMS. Plasma concentrations following non-(14)C-labelled oral therapeutic doses were measured using either HPLC-electrochemical detection (clarithromycin) or HPLC-UV (sumatriptan, propafenone). For all five drugs an oral microdose predicted reasonably well the PK, including the shape of the plasma profile, following an oral therapeutic dose. For clarithromycin, sumatriptan, and propafenone, one parameter, oral bioavailability, was marginally outside of the normally acceptable 2-fold prediction interval around the mean therapeutic dose value. For clarithromycin, sumatriptan and propafenone, data obtained from an oral and iv microdose were compared within the same cohort of subjects used in the study, as well as those reported in the literature. For paracetamol (oral and iv) and phenobarbital (oral), microdose data were compared with those reported in the literature only. Where 100 µg iv (14)C-doses were given alone and with an oral non-labelled therapeutic dose, excellent accord between the PK parameters was observed indicating that the disposition kinetics of the drugs tested were unaffected by the presence of therapeutic concentrations. This observation implies that any deviation from linearity following the oral therapeutic doses occurs during the absorption process.


Subject(s)
Acetaminophen , Clarithromycin , Phenobarbital , Propafenone , Sumatriptan , Acetaminophen/administration & dosage , Acetaminophen/blood , Acetaminophen/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Carbon Radioisotopes/administration & dosage , Carbon Radioisotopes/blood , Carbon Radioisotopes/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Clarithromycin/administration & dosage , Clarithromycin/blood , Clarithromycin/pharmacokinetics , Cross-Over Studies , Dose-Response Relationship, Drug , Humans , Injections, Intravenous , Male , Mass Spectrometry/methods , Middle Aged , Phenobarbital/administration & dosage , Phenobarbital/blood , Phenobarbital/pharmacokinetics , Propafenone/administration & dosage , Propafenone/blood , Propafenone/pharmacokinetics , Sumatriptan/administration & dosage , Sumatriptan/blood , Sumatriptan/pharmacokinetics
13.
Drug Metab Dispos ; 38(12): 2139-46, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20847136

ABSTRACT

Accurate assignment of the concentration of victim drug/inhibitor available at the enzyme active site, both in vivo and within an in vitro incubation, is an essential requirement in rationalizing and predicting drug-drug interactions. Inhibitor accumulation within the liver, whether as a result of active transport processes or intracellular binding, may best be accounted for using hepatocytes rather than hepatic microsomes to estimate in vitro inhibitory potency. The aims of this study were to compare K(i) values determined in rat liver microsomes and freshly isolated rat hepatocytes of four cytochrome P450 (P450) inhibitors (clarithromycin, enoxacin, nelfinavir, and saquinavir) with known hepatic transporter involvement and a range of uptake (cell/medium concentration ratios 20-3000) and clearance (10-1200 µl/min/10(6) cells) properties. Inhibition studies were performed using two well established P450 probe substrates (theophylline and midazolam). Comparison of unbound K(i) values showed marked differences between the two in vitro systems for inhibition of metabolism. In two cases (clarithromycin and enoxacin, both low-clearance drugs), inhibitory potency in hepatocytes markedly exceeded that in microsomes (10- to 20-fold), and this result was consistent with their high cell/medium concentration ratios. For nelfinavir and saquinavir (high-clearance, extensively metabolized drugs), the opposite trend was seen in the K(i) values: despite very high cell/medium concentration ratios, stronger inhibition was evident within microsomal preparations. Hence, the consequences of hepatic accumulation resulting from uptake transporters vary according to the clearance of the inhibitor. This study demonstrates that transporter-enzyme interplay can result in differences in inhibitory potency between microsomes and hepatocytes and hence drug-drug interaction predictions that are not always intuitive.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Hepatocytes/enzymology , Microsomes, Liver/enzymology , Animals , Cell Separation , Clarithromycin/pharmacology , Drug Interactions , Enoxacin/pharmacology , Male , Midazolam/metabolism , Nelfinavir/pharmacology , Rats , Rats, Sprague-Dawley , Saquinavir/pharmacology , Theophylline/metabolism
14.
Stem Cell Res ; 5(1): 4-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20483202

ABSTRACT

Hepatotoxicity is an enormous and increasing problem for the pharmaceutical industry. Early detection of problems during the drug discovery pathway is advantageous to minimize costs and improve patient safety. However, current cellular models are sub-optimal. This review addresses the potential use of pluripotent stem cells in the generation of hepatic cell lineages. It begins by highlighting the scale of the problem faced by the pharmaceutical industry, the precise nature of drug-induced liver injury and where in the drug discovery pathway the need for additional cell models arises. Current research is discussed, mainly for generating hepatocyte-like cells rather than other liver cell-types. In addition, an effort is made to identify where some of the major barriers remain in translating what is currently hypothesis-driven laboratory research into meaningful platform technologies for the pharmaceutical industry.


Subject(s)
Cell Culture Techniques/methods , Cell Lineage , Liver/cytology , Liver/drug effects , Pluripotent Stem Cells/cytology , Animals , Drug Evaluation, Preclinical , Drug Industry , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...