Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-37003144

ABSTRACT

BACKGROUND: Aspirin and eicosapentaenoic acid (EPA) have colorectal polyp prevention activity, alone and in combination. This study measured levels of plasma and rectal mucosal oxylipins in participants of the seAFOod 2 × 2 factorial, randomised, placebo-controlled trial, who received aspirin 300 mg daily and EPA 2000 mg free fatty acid, alone and in combination, for 12 months. METHODS: Resolvin (Rv) E1, 15-epi-lipoxin (LX) A4 and respective precursors 18-HEPE and 15-HETE (with chiral separation) were measured by ultra-high performance liquid chromatography-tandem mass spectrometry in plasma taken at baseline, 6 months and 12 months, as well as rectal mucosa obtained at trial exit colonoscopy at 12 months, in 401 trial participants. RESULTS: Despite detection of S- and R- enantiomers of 18-HEPE and 15-HETE in ng/ml concentrations, RvE1 or 15­epi-LXA4 were not detected above a limit of detection of 20 pg/ml in plasma or rectal mucosa, even in individuals randomised to both aspirin and EPA. We have confirmed in a large clinical trial cohort that prolonged (12 months) treatment with EPA is associated with increased plasma 18-HEPE concentrations (median [inter-quartile range] total 18-HEPE 0.51 [0.21-1.95] ng/ml at baseline versus 0.95 [0.46-4.06] ng/ml at 6 months [P<0.0001] in those randomised to EPA alone), which correlate strongly with respective rectal mucosal 18-HEPE levels (r = 0.82; P<0.001), but which do not predict polyp prevention efficacy by EPA or aspirin. CONCLUSION: Analysis of seAFOod trial plasma and rectal mucosal samples has not provided evidence of synthesis of the EPA-derived specialised pro-resolving mediator RvE1 or aspirin-trigged lipoxin 15­epi-LXA4. We cannot rule out degradation of individual oxylipins during sample collection and storage but readily measurable precursor oxylipins argues against widespread degradation.


Subject(s)
Aspirin , Lipoxins , Humans , Aspirin/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Oxylipins , Mucous Membrane
2.
J Pharmacol Exp Ther ; 370(2): 242-251, 2019 08.
Article in English | MEDLINE | ID: mdl-31189729

ABSTRACT

Adequate drug distribution through tumors is essential for treatment to be effective. Palbociclib is a cyclin-dependent kinase 4/6 inhibitor approved for use in patients with hormone receptor positive, human epidermal growth factor receptor 2 negative metastatic breast cancer. It has unusual physicochemical properties, which may significantly influence its distribution in tumor tissue. We studied the penetration and distribution of palbociclib in vitro, including the use of multicellular three-dimensional models and mathematical modeling. MCF-7 and DLD-1 cell lines were grown as single cell suspensions (SCS) and spheroids; palbociclib uptake and efflux were studied using liquid chromatography-tandem mass spectrometry. Intracellular concentrations of palbociclib for MCF-7 SCS (C max 3.22 µM) and spheroids (C max 2.91 µM) were 32- and 29-fold higher and in DLD-1, 13- and 7-fold higher, respectively, than the media concentration (0.1 µM). Total palbociclib uptake was lower in DLD-1 cells than MCF-7 cells in both SCS and spheroids. Both uptake and efflux of palbociclib were slower in spheroids than SCS. These data were used to develop a mathematical model of palbociclib transport that quantifies key parameters determining drug penetration and distribution. The model reproduced qualitatively most features of the experimental data and distinguished between SCS and spheroids, providing additional support for hypotheses derived from the experimental data. Mathematical modeling has the potential for translating in vitro data into clinically relevant estimates of tumor drug concentrations. SIGNIFICANCE STATEMENT: This study explores palbociclib uptake and efflux in single cell suspension and spheroid models of cancer. Large intracellular concentrations of palbociclib are found after drug exposure. The data from this study may aid understanding of the intratumoural pharmacokinetics of palbociclib, which is useful in understanding how drug distributes within tumor tissue and optimizing drug efficacy. Biomathematical modelling has the potential to derive intratumoural drug concentrations from plasma pharmacokinetics in patients.


Subject(s)
Piperazines/metabolism , Pyridines/metabolism , Spheroids, Cellular/metabolism , Biological Transport , Cell Survival/drug effects , Humans , MCF-7 Cells , Models, Biological , Piperazines/pharmacology , Pyridines/pharmacology , Single-Cell Analysis , Spheroids, Cellular/drug effects
3.
R Soc Open Sci ; 4(5): 170014, 2017 May.
Article in English | MEDLINE | ID: mdl-28573005

ABSTRACT

The tumour vasculature and microenvironment is complex and heterogeneous, contributing to reduced delivery of cancer drugs to the tumour. We have developed an in silico model of drug transport in a tumour cord to explore the effect of different drug regimes over a 72 h period and how changes in pharmacokinetic parameters affect tumour exposure to the cytotoxic drug doxorubicin. We used the model to describe the radial and axial distribution of drug in the tumour cord as a function of changes in the transport rate across the cell membrane, blood vessel and intercellular permeability, flow rate, and the binding and unbinding ratio of drug within the cancer cells. We explored how changes in these parameters may affect cellular exposure to drug. The model demonstrates the extent to which distance from the supplying vessel influences drug levels and the effect of dosing schedule in relation to saturation of drug-binding sites. It also shows the likely impact on drug distribution of the aberrant vasculature seen within tumours. The model can be adapted for other drugs and extended to include other parameters. The analysis confirms that computational models can play a role in understanding novel cancer therapies to optimize drug administration and delivery.

4.
J R Soc Interface ; 11(94): 20131173, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24621814

ABSTRACT

The ability to predict how far a drug will penetrate into the tumour microenvironment within its pharmacokinetic (PK) lifespan would provide valuable information about therapeutic response. As the PK profile is directly related to the route and schedule of drug administration, an in silico tool that can predict the drug administration schedule that results in optimal drug delivery to tumours would streamline clinical trial design. This paper investigates the application of mathematical and computational modelling techniques to help improve our understanding of the fundamental mechanisms underlying drug delivery, and compares the performance of a simple model with more complex approaches. Three models of drug transport are developed, all based on the same drug binding model and parametrized by bespoke in vitro experiments. Their predictions, compared for a 'tumour cord' geometry, are qualitatively and quantitatively similar. We assess the effect of varying the PK profile of the supplied drug, and the binding affinity of the drug to tumour cells, on the concentration of drug reaching cells and the accumulated exposure of cells to drug at arbitrary distances from a supplying blood vessel. This is a contribution towards developing a useful drug transport modelling tool for informing strategies for the treatment of tumour cells which are 'pharmacokinetically resistant' to chemotherapeutic strategies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Models, Biological , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Biological Transport , Computer Simulation , Humans
5.
Br J Pharmacol ; 166(5): 1724-37, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22300262

ABSTRACT

BACKGROUND AND PURPOSE: The omega-3 polyunsaturated fatty acid (PUFA) eicosapentaenoic acid (EPA) has antineoplastic activity at early stages of colorectal carcinogenesis, relevant to chemoprevention of colorectal cancer (CRC). We tested the hypothesis that EPA also has anti-CRC activity at later stages of colorectal carcinogenesis, relevant to treatment of metastatic CRC, via modulation of E-type PG synthesis. EXPERIMENTAL APPROACH: A BALB/c mouse model, in which intrasplenic injection of syngeneic MC-26 mouse CRC cells leads to development of liver metastases, was used. Dietary EPA was administered in the free fatty acid (FFA) form for 2 weeks before and after ultrasound-guided intrasplenic injection of 1 × 10(6) MC-26 cells (n= 16 each group). KEY RESULTS: Treatment with 5% (w w(-1)) EPA-FFA was associated with a reduced MC-26 mouse CRC cell liver tumour burden compared with control animals (median liver weight 1.03 g vs. 1.62 g; P < 0.034). Administration of 5% EPA-FFA was also linked to a significant increase in tumour EPA incorporation and lower intratumoural PGE(2) levels (with concomitant increased production of PGE(3)). Liver tumours from 5% EPA-FFA- treated mice demonstrated decreased 5-bromo-2-deoxyuridine-positive CRC cell proliferation and reduced phosphorylated ERK 1/2 expression at the invasive edge of tumours. A concentration-dependent reduction in MC-26 CRC cell Transwell® migration following EPA-FFA treatment (50-200 µM) in vitro was rescued by exogenous PGE(2) (10 µM) and PGE(1)-alcohol (1 µM). CONCLUSIONS AND IMPLICATIONS: EPA-FFA inhibits MC-26 CRC cell liver metastasis. EPA incorporation is associated with a 'PGE(2) to PGE(3) switch' in liver tumours. Inhibition of PGE(2)-EP(4) receptor-dependent CRC cell motility probably contributes to the antineoplastic activity of EPA.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Dinoprostone/metabolism , Eicosapentaenoic Acid/therapeutic use , Liver Neoplasms/drug therapy , Alprostadil/analogs & derivatives , Alprostadil/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclooxygenase 2/metabolism , Eicosapentaenoic Acid/pharmacology , Female , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Tumor Burden/drug effects
6.
J Theor Biol ; 257(4): 598-608, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19183560

ABSTRACT

Inadequate drug delivery to tumours is now recognised as a key factor that limits the efficacy of anticancer drugs. Extravasation and penetration of therapeutic agents through avascular tissue are critically important processes if sufficient drug is to be delivered to be therapeutic. The purpose of this study is to develop an in silico model that will simulate the transport of the clinically used cytotoxic drug doxorubicin across multicell layers (MCLs) in vitro. Three cell lines were employed: DLD1 (human colon carcinoma), MCF7 (human breast carcinoma) and NCI/ADR-Res (doxorubicin resistant and P-glycoprotein [Pgp] overexpressing ovarian cell line). Cells were cultured on transwell culture inserts to various thicknesses and doxorubicin at various concentrations (100 or 50 microM) was added to the top chamber. The concentration of drug appearing in the bottom chamber was determined as a function of time by HPLC-MS/MS. The rate of drug penetration was inversely proportional to the thickness of the MCL. The rate and extent of doxorubicin penetration was no different in the presence of NCI/ADR-Res cells expressing Pgp compared to MCF7 cells. A mathematical model based upon the premise that the transport of doxorubicin across cell membrane bilayers occurs by a passive "flip-flop" mechanism of the drug between two membrane leaflets was constructed. The mathematical model treats the transwell apparatus as a series of compartments and the MCL is treated as a series of cell layers, separated by small intercellular spaces. This model demonstrates good agreement between predicted and actual drug penetration in vitro and may be applied to the prediction of drug transport in vivo, potentially becoming a useful tool in the study of optimal chemotherapy regimes.


Subject(s)
Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/pharmacokinetics , Models, Biological , Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/administration & dosage , Biological Transport , Cell Membrane/metabolism , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Drug Resistance, Neoplasm , Female , Humans , Neoplasm Proteins/metabolism , Tumor Cells, Cultured
7.
Eur J Cancer ; 43(11): 1764-71, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17600697

ABSTRACT

Improved understanding of the involvement of matrix metalloproteinases (MMPs), including membrane-type MMPs (MT-MMPs), in human tumours has potential diagnostic, prognostic and therapeutic implications. We assessed the relationship between MT-MMP expression and clinicopathological parameters in human non-small cell lung cancer (NSCLC) and histologically normal lung tissue by quantitative Real Time PCR (qRT-PCR). All MT-MMPs (MMPs 14-17, 24 and 25) were detected by qRT-PCR with significantly higher MMP-14, -15 and -17 expression observed in tumour relative to normal lung specimens. MMP-16 was undetectable in normal lung but expressed in 8% tumours. MMP-15 demonstrated significant overexpression in adenocarcinomas relative to squamous cell carcinomas and normal lung tissue. MMP-14 mRNA expression strongly correlated to MMP-14 proteolytic activity in preclinical tumour models, indicating that qRT-PCR may predict MMP-14 activity levels in NSCLC. These data suggest that MMP-14, -15 and -17 may be good markers of disease, or therapeutic targets for treatment of human NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Matrix Metalloproteinases/metabolism , Neoplasm Proteins/metabolism , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Inbred Strains , Middle Aged , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Transplantation, Heterologous
8.
Ann Oncol ; 18(6): 1098-103, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17442658

ABSTRACT

BACKGROUND: AQ4N (1,4-bis[[2-(dimethylamino)ethyl] amino]-5,8-dihydroxyanthracene-9, 10-dione bis-N-oxide dihydrochloride) is a prodrug which is selectively activated within hypoxic tissues to AQ4, a topoisomerase II inhibitor and DNA intercalator. PATIENTS AND METHODS: In the phase I study, 22 patients with oesophageal carcinoma received an i.v. infusion of AQ4N (22.5-447 mg/m(2)) followed, 2 weeks later, by further infusion and radiotherapy. Pharmacokinetics and lymphocyte AQ4N and AQ4 levels were measured after the first dose. At 447 mg/m(2), biopsies of tumour and normal tissue were taken after AQ4N administration. RESULTS: Drug-related adverse events were blue discolouration of skin and urine, grade 2-3 lymphopenia, grade 1-3 fatigue, grade 1-2 anaemia, leucopenia and nausea. There were no drug-related serious adverse events (SAEs). Three patients had reductions in tumour volume >50%, nine had stable disease. Pharmacokinetics indicated predictable clearance. Plasma area under the curve (AUC) at 447 mg/m(2) exceeded AQ4N concentrations in mice at therapeutic doses and tumour biopsies contained concentrations of AQ4 greater than those in normal tissue. Tumour concentrations of AQ4 exceeded in vitro IC(50) values for most cell lines investigated. CONCLUSIONS: No dose-limiting toxic effects were observed and a maximum tolerated dose was not established. Tumour AQ4 concentrations and plasma AUC at 447 mg/m(2) exceeded active levels in preclinical models. This dose was chosen for future studies with radiotherapy.


Subject(s)
Anthraquinones/pharmacokinetics , Anthraquinones/toxicity , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/radiotherapy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Aged , Aged, 80 and over , Anthraquinones/administration & dosage , Area Under Curve , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Combined Modality Therapy , Female , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Patient Selection
9.
J Chemother ; 19(1): 66-78, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17309854

ABSTRACT

The pyrrolobenzodiazepine monomer DRH-417 is a member of the anthramycin group of anti-tumor antibiotics that bind covalently to the N2 of guanine within the minor groove of DNA. DRH-417 emerged from the EORTC-Drug Discovery Committee and NCI 60 cell line in vitro screening programs as a potent antiproliferative agent with differential sensitivity towards certain cancer types such as melanoma, breast and renal cell carcinoma (mean IC(50) = 3 nM). DRH-417 was therefore tested for in vivo activity. The maximum tolerated dose (MTD) was established as 0.5 mg/kg given i.p. Marked anti-tumor activity was seen in two human renal cell cancers, one breast cancer and a murine colon tumor model (p<0.01). A selective HPLC (LC/MS) analytical method was developed and plasma pharmacokinetics determined. At a dose of 0.5 mg kg(-1), the plasma AUC was 540 nM h (197.1 ng h ml(-1)) and the peak plasma concentration (171 nM [62.4 ng ml(-1)]) occurred at 30 min., reaching doses levels well above those needed for in vitro antiproliferative activity. Genomic profiling of in vivo sensitive tumors revealed that the latter have an activated insulin-like growth factor signaling pathway.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Benzodiazepines/pharmacology , Pyrroles/pharmacology , Animals , Anthramycin/pharmacology , Antibiotics, Antineoplastic/analysis , Antibiotics, Antineoplastic/pharmacokinetics , Benzodiazepines/analysis , Benzodiazepines/therapeutic use , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Gene Expression Profiling , Humans , Mass Spectrometry , Mice , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Pyrroles/analysis , Pyrroles/therapeutic use , Transplantation, Heterologous
10.
Teratog Carcinog Mutagen ; Suppl 2: 13-29, 2003.
Article in English | MEDLINE | ID: mdl-14691977

ABSTRACT

In order to determine differences in repair after treatment with DNA damaging agents, normal and cancer cells were selected for analysis of single strand breaks and DNA crosslinks using the Comet assay. Normal human lymphocytes, human colorectal adenocarcinoma SW620 cells, lung carcinoma A549, and H460 cell lines were exposed to an ethylating agent (ethylmethane sulfonate [EMS]), and a cross-linking agent (mitomycin C [MMC]). Differences in repair profiles of DNA damage demonstrated using the comet assay were observed in human lymphocytes and tumour cell lines with both mutagens. Results were also indicative that MMC repair is concentration-dependent. It was also apparent that normal cells repair DNA damage more readily than tumour cells. Repair also varied between different cell lines. To investigate the mechanistic differences of these two chemicals, flow cytometry studies were undertaken in tumour cells, namely cell cycle analysis and frequency of micronuclei induction (FMN). A G2M phase block was clearly evident following treatment with EMS at all concentrations tested. With MMC, an initial arrest of cells in G2M was accompanied by a build-up in S-phase over longer exposure periods. Also, at the highest mutagen doses there were different patterns of micronuclei induction. Thus, using the mutagens with different mechanisms of action highlighted the differences in repair patterns between normal and tumour cells.


Subject(s)
Comet Assay/methods , DNA Repair , DNA, Neoplasm/drug effects , DNA/drug effects , Flow Cytometry/methods , Mutagens/toxicity , Cell Cycle/drug effects , Cells, Cultured , Cross-Linking Reagents/toxicity , DNA Damage , Ethyl Methanesulfonate/toxicity , Humans , Lymphocytes/drug effects , Micronuclei, Chromosome-Defective/drug effects , Mitomycin/toxicity , Neoplasms/genetics
11.
Br J Pharmacol ; 137(5): 701-9, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12381684

ABSTRACT

The bioreductive drug EO9 (3-hydroxy-5-aziridinyl-1-methyl-2[indole-4,7-dione]-prop-beta-en-alpha-ol) has good pharmacodynamic properties in vitro, modest anti-tumour activity in experimental tumour models, but failed to show activity in clinical trials. Understanding the reasons for its poor efficacy in vivo is important in terms of progressing second generation analogues into the clinic. In two human tumour xenografts, direct intra-tumoural injection resulted in improved anti-tumour activity compared with intravenous administration suggesting that drug delivery to tumours is suboptimal. Compared with Mitomycin C (MMC) and the experimental agent MeDZQ, EO9 was rapidly cleared from the systemic circulation (t1/2=1.8 min) whereas MMC and MeDZQ had significantly increased plasma t1/2 values (14 and 22 min respectively). These three compounds demonstrated similar pharmacodynamic properties in terms of potency towards the NQO1 (NAD(P)H:Quinone oxidoreductase) rich H460 cell line in vitro but differed significantly in their in vivo activity with growth delays of 17.7, 4.5 and 1.0 days for MMC, MeDZQ and EO9 respectively. EO9 was rapidly metabolized by red blood cells in vitro (t1/2=14.5 min) which must contribute to its rapid pharmacokinetic elimination in vivo whereas MMC and MeDZQ were metabolized at comparatively slower rates (t1/2>120 min and 77.0 min respectively). In conclusion, the development of second generation EO9 analogues should address the issue of drug delivery and analysis of drug metabolism by murine whole blood in vitro could be utilized as a preliminary screen to identify lead compounds that are likely to have improved pharmacokinetic profiles in vivo.


Subject(s)
Aziridines/administration & dosage , Aziridines/chemistry , Indolequinones , Indoles/administration & dosage , Indoles/chemistry , Quinones/administration & dosage , Technology, Pharmaceutical/methods , Animals , Aziridines/blood , Drug Delivery Systems/methods , Humans , Indoles/blood , Mice , Mice, Nude , Quinones/blood , Quinones/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays/methods
12.
Drug Metab Dispos ; 29(4 Pt 1): 422-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259326

ABSTRACT

AQ4N (1,4-bis-[[2-(dimethylamino-N-oxide)ethyl]amino]5,8-dihydroxyanthracene-9,10-dione) is in a class of bioreductive agents incorporating the aliphatic N-oxide functionality and is well documented as a very effective enhancer of radiotherapy and chemotherapy. The compound is shortly to enter Phase I clinical trials in the United Kingdom, and this study describes the preclinical pharmacokinetics and metabolism of AQ4N in mice. AQ4N was administered by i.v. injection at doses of 200, 100, and 20 mg/kg and was quantified by high-performance liquid chromatography and liquid chromatography/mass spectroscopy. There was a linear increase in the maximum plasma concentration (Cmax) proportional to dose with a Cmax of 1171 microg/ml at the maximum tolerated dose of 200 mg/kg. The area under plasma concentration versus time curve (AUC) increased disproportionately with dose from 14.1 microg/h/ml at 20 mg/kg to 247 microg/h/ml at 200 mg/kg with a subsequent decrease in clearance. Terminal elimination half-lives ranged from 0.64 to 0.83 h. The spectra of the two major metabolites matched those from authentic standards with the molecular ions [M + H]+ being detected at m/z 445.4 (AQ4N), m/z 429.5 (AQ4 mono-N-oxide) and m/z 413.5 (AQ4). Only low concentrations of the toxic metabolite (AQ4) were detected in plasma at all three doses, with the AUC and Cmax at 200 mg/kg being 3.54 microg/h/ml and 3.7 microg/ml, respectively, representing <2% of AQ4N. Concentrations of the intermediate AQ4 M represented 8, 10, and 18% of those for AQ4N at the doses of 20,100, and 200 mg/kg. The concentrations necessary for a therapeutic response in vivo have been described in this pharmacokinetic study.


Subject(s)
Anthraquinones/pharmacokinetics , Prodrugs/pharmacokinetics , Animals , Anthraquinones/metabolism , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Female , Mice , Prodrugs/metabolism , Radiation-Protective Agents/metabolism , Radiation-Protective Agents/pharmacokinetics
13.
J Chromatogr B Biomed Sci Appl ; 764(1-2): 193-206, 2001 Nov 25.
Article in English | MEDLINE | ID: mdl-11817028

ABSTRACT

The quinoid anthracycline-related anti-cancer agents represent an important group of anti-tumour drugs with a wide spectrum of activity. We review here some of the separation techniques used for the analysis of anthracyclines and related compounds. In this review we have covered a range of compounds from the early anthracycline antibiotics such as doxorubicin to the more recent anthracenediones and anthrapyrazoles such as mitoxantrone and losoxantrone, respectively. We also include novel compounds such as AQ4N and C1311, both awaiting clinical trial. Separations of the anthraquinone related anti-cancer agents are predominantly by HPLC. These separation techniques have been used for a variety of applications including drug stability, protein binding and therapeutic drug monitoring as well as detailed pharmacokinetic and metabolic studies. Pharmacokinetics, and therefore drug analysis, plays a central role in both the development of new agents and also leads to a better understanding of clinically established agents in this class. Sample preparation and extraction methods including solid-phase and liquid-liquid extraction have also been highlighted. Many anthraquinone related compounds are highly coloured and fluoresce. They are suitable for a range of detection methods including UV-Vis, electrochemical and fluorescence. The methods described are used for sometimes complex separations that are needed for the evaluation of such compounds in biological samples.


Subject(s)
Anthraquinones/isolation & purification , Antineoplastic Agents/isolation & purification , Chromatography, High Pressure Liquid/methods
14.
Cancer Res ; 60(22): 6384-90, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103802

ABSTRACT

Mitomycin C (MMC) is a clinically used anticancer drug that is reduced to cytotoxic metabolites by cellular reductases via a process known as bioreductive drug activation. The identification of key enzymes responsible for drug activation has been investigated extensively with the ultimate aim of tailoring drug administration to patients whose tumors possess the biochemical machinery required for drug activation. In the case of MMC, considerable interest has been centered upon the enzyme DT-diaphorase (DTD) although conflicting reports of good and poor correlations between enzyme activity and response in vitro and in vivo have been published. The principle aim of this study was to provide a definitive answer to the question of whether tumor response to MMC could be predicted on the basis of DTD activity in a large panel of human tumor xenografts. DTD levels were measured in 45 human tumor xenografts that had been characterized previously in terms of their sensitivity to MMC in vitro and in vivo (the in vivo response profile to MMC was taken from work published previously). A poor correlation between DTD activity and antitumor activity in vitro as well as in vivo was obtained. This study also assessed the predictive value of an alternative approach based upon the ability of tumor homogenates to metabolize MMC. This approach is based on the premise that the overall rate of MMC metabolism may provide a better indicator of response than single enzyme measurements. MMC metabolism was evaluated in tumor homogenates (clarified by centrifugation at 1000 x g for 1 min) by measuring the disappearance of the parent compound by HPLC. In responsive [T/C <10% (T/C defined as the relative size of treated and control tumors)] and resistant (T/C >50%) tumors, the mean half life of MMC was 75+/-48.3 and 280+/-129.6 min, respectively. The difference between the two groups was statistically significant (P < 0.005). In conclusion, these results unequivocally demonstrate that response to MMC in vivo cannot be predicted on the basis of DTD activity. Measurement of MMC metabolism by tumor homogenates on the other hand may provide a better indicator of tumor response, and further studies are required to determine whether this approach has real clinical potential in terms of individualizing patient chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Mitomycin/metabolism , Mitomycin/pharmacology , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasms/drug therapy , Neoplasms/enzymology , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Biotransformation , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Mitomycin/pharmacokinetics , Predictive Value of Tests , Xenograft Model Antitumor Assays
15.
J Chromatogr B Biomed Sci Appl ; 742(2): 239-45, 2000 Jun 09.
Article in English | MEDLINE | ID: mdl-10901128

ABSTRACT

A simple, highly selective and reproducible reversed-phase high-performance liquid chromatography method has been developed for the analysis of the new anti-cancer pro-drug AQ4N. The sample pre-treatment involves a simple protein precipitation protocol, using methanol. Chromatographic separations were performed using a HiChrom HIRPB (25 cmX4.6 mm I.D.) column, with mobile phase of acetonitrile-ammonium formate buffer (0.05 M) (22:78, v/v), with final pH adjusted to 3.6 with formic acid. The flow-rate was maintained at 1.2 ml min(-1). Detection was via photodiode array performed in the UV range at 242 nm and, since the compounds are an intense blue colour, in the visible range at 612 nm. The structurally related compound mitoxantrone was used as internal standard. The validated quantification range of the method was 0.05-10.0 microg ml(-1) in mouse plasma. The inter-day relative standard deviations (RSDs) (n=5) ranged from 18.4% and 12.1% at 0.05 microg ml(-1) to 2.9% and 3.3% at 10.0 microg ml(-1) for AQ4N and AQ4, respectively. The intra-day RSDs for supplemented mouse plasma (n=6) ranged from 8.2% and 14.2% at 0.05 microg ml(-1) to 7.6% and 11.5% at 10.0 microg ml(-1) for AQ4N and AQ4, respectively. The overall recovery of the procedure for AQ4N was 89.4 +/- 1.77% and 76.1 +/- 7.26% for AQ4. The limit of detection was 50 ng ml(-1) with a 100 microl sample volume. The method described provides a suitable technique for the future analysis of low levels of AQ4N and AQ4 in clinical samples.


Subject(s)
Anthraquinones/blood , Antineoplastic Agents/blood , Chromatography, High Pressure Liquid/methods , Prodrugs/pharmacokinetics , Animals , Anthraquinones/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Blood Proteins/metabolism , Calibration , Female , Mice , Protein Binding , Reference Standards
16.
Anticancer Res ; 20(1A): 229-33, 2000.
Article in English | MEDLINE | ID: mdl-10769660

ABSTRACT

The di-sodium phosphate pro-drug of combretastatin-A4(combA-4P) is undergoing Phase 1 clinical trial in the USA and UK. Its mechanism of action is thought to be related to tubulin-binding properties that result in rapid, tumour endothelial cell damage, neovascular shutdown and subsequent haemorrhagic necrosis. Drugs that work by this mechanism are unlikely to eradicate the tumour as a single agent but should potentiate standard chemotherapy. This study demonstrates that extensive necrosis occurred in a treated refractory murine colon adenocarcinoma but the damage was not accompanied by any measurable effect on tumour growth. Tumours continued to grow from the viable rim that remained. Combination chemotherapy with 5-fluorouracil (5-FU) resulted in significant (p < 0.01) anti-tumour effects. Measurement of 5-FU concentrations suggested that this was true synergism and not simply a pharmacokinetic interaction due to the vascular mechanism of combA-4P. The study suggests that if an antivascular mechanism can be demonstrated in humans, combination chemotherapy should be rapidly assessed in a clinical setting.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Adenocarcinoma/blood supply , Adenocarcinoma/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols/blood , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Drug Synergism , Endothelium, Vascular/drug effects , Female , Fluorouracil/administration & dosage , Fluorouracil/blood , Fluorouracil/pharmacokinetics , Mice , Necrosis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Transplantation , Prodrugs/administration & dosage , Prodrugs/pharmacokinetics , Stilbenes/administration & dosage , Stilbenes/blood , Stilbenes/pharmacokinetics , Tubulin Modulators
17.
Biochem Pharmacol ; 59(7): 831-7, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10718341

ABSTRACT

RH1 (2,5-diaziridinyl-3-(hydroxylmethyl)-6-methyl-1,4-benzoquinone) has shown preferential activity against human tumour cell lines which express high levels of DTD (EC 1.6.99.2; NAD(P)H:quinone oxidoreductase, NQO1, DT-diaphorase) and is a candidate for clinical trials. EO9 (3-hydroxy-5-aziridinyl-1-methyl-2-[1H indole-4,7-dione]prop-beta-en-alpha-ol) is a known substrate for DTD but clinical trials were disappointing, as a result of rapid plasma clearance and reversible dose-limiting kidney toxicity. It is an obvious concern that RH1 does not exhibit the same limitations. We therefore describe the antitumour activity and pharmacology of RH1 in mice and compare its pharmacological characteristics to those of EO9. Significant antitumour activity (P = 0.01) was seen for RH1 (0.5 mg/kg, i.p.) against the high DTD-expressing H460 human lung carcinoma. Pharmacokinetic analysis of RH1 in mice showed a t1/2 of 23 min with an area under the curve of 43.0 ng hr mL(-1) resulting in a calculated clearance of 5.1 mL min(-1), 10-fold slower than EO9. RH1 was also more stable than EO9 in murine blood, where the breakdown was thought to be DTD-related. NADH-dependent microsomal metabolism of RH1 and EO9 in both liver and kidney was slow (<100 pmol/min/g tissue), reflecting the low microsomal DTD expression (<35 nmol/mg/min). Liver cytosol metabolism was rapid for both compounds (>4500 pmol/min/g tissue), although DTD levels were low (21.4+/-0.6 nmol/mg/min). DTD activity in the kidney cytosol was high (125+/-8.2 nmol/mg/min) and EO9 was rapidly metabolised (4396+/-1678 pmol/min/g), but the metabolic rate for RH1 was 7-fold slower (608+/-86 pmol/min/g), even though RH1 was shown to be an excellent substrate for DTD (Vmax = 800 micromol/min/mg and a Km of 11.8 microM). The two DTD substrates RH1 and EO9 are clearly metabolised differently, suggesting that RH1 may have different pharmacological properties to those of EO9 in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Aziridines/pharmacology , Benzoquinones/pharmacology , Indolequinones , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Aziridines/pharmacokinetics , Aziridines/therapeutic use , Benzoquinones/pharmacokinetics , Benzoquinones/therapeutic use , Drug Screening Assays, Antitumor , Drug Stability , Humans , Indoles/pharmacokinetics , Indoles/pharmacology , Kinetics , Lung Neoplasms/metabolism , Mice , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasm Transplantation
18.
Clin Cancer Res ; 5(11): 3682-8, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10589787

ABSTRACT

PK1 is a synthetic N-(2-hydroxypropyl)methacrylamide copolymer-doxorubicin (dox) conjugate currently undergoing Phase II evaluation in the United Kingdom. We have studied the activity of PK1 in three murine colon tumor models that differ in terms of morphology and vascularization in an attempt to determine which factors are most important in the tumor response to PK1. Vascular permeability was evaluated with Evans Blue, and pharmacokinetic studies in MAC15A and MAC26 used high-performance liquid chromatography to monitor both PK1 uptake and dox release in the tumors. Cathepsin B activity was assessed using a specific substrate. PK1 (40 mg x kg(-1) dox equivalent) was significantly more effective than dox alone (10 mg x kg(-1)) was against MAC15A tumors, which possess enhanced perfusion and retention, but not against MAC26 tumors, although MAC15A was also responsive to PK1 when grown as avascular micrometastatic deposits in the lung. Pharmacokinetic studies showed similar levels of PK1 in both tumors. Peak tumor levels of released dox were 7-fold greater in the responsive MAC15A tumor (53 microg x ml(-1)) compared with the less responsive MAC26 tumor (7.7 microg x ml(-1)) and more than 18-fold greater in MAC15A than when free dox was given. These differences in response correlated also with an increased lysosomal activity of cathepsin B. Calculated AUCs for intratumoral dox released were 431 microg x h x g(-1) and 775 microg x h x g(-1) for MAC15A and MAC26, respectively. These AUCs are 4-fold and 7-fold higher, respectively, than when dox is given alone. This study has shown that activity and the pharmacokinetics of PK1 and released dox are dependent on both the vascular properties and enzyme content of the tumors. These studies are likely to have clinical implications as aggressive tumors are known to have increased protease activity.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Colonic Neoplasms/drug therapy , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Polymethacrylic Acids/pharmacokinetics , Animals , Antineoplastic Agents/therapeutic use , Area Under Curve , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Carriers , Female , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Mice , Mice, Inbred Strains , Polymethacrylic Acids/administration & dosage , Polymethacrylic Acids/therapeutic use , Tissue Distribution
19.
Drug Metab Dispos ; 27(2): 240-5, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9929509

ABSTRACT

C1311 has emerged as the lead compound from a novel group of anticancer agents, the imidazoacridinones, and will be entering clinical trials shortly. Previous murine pharmacokinetic studies have shown C1311 to be rapidly and extensively distributed into tissues including tumor. This study has identified two major metabolites of C1311 and describes their pharmacokinetics in mice. M1 is a glucuronide of the parent compound with high concentrations in both plasma and liver. Calculated area under the plasma concentration versus time curve values were 6-fold and 2-fold greater, respectively, than C1311. Based on these studies, we propose M2 to be a nonfluorescent oxidation product because electrospray ionization-mass spectroscopy/mass spectroscopy analysis gave a molecular ion at m/z 367, 16 U greater than the parent compound. It formed rapidly in liver preparations in vitro, both murine and human, by a cytosolic process in the presence of NADPH and in vivo was detected in liver tissues at concentrations equivalent to those of C1311 but was not detectable in plasma. Preliminary in vitro toxicity studies showed M2 to be as potent as C1311 against MAC15A tumor cells. Over the first 24 h, 39% of the administered dose is eliminated via the bile (28%) mostly as C1311 or the kidneys (11%) as the glucuronide (M1). This study has given valuable information as to the likely metabolic pathway to occur in humans, and the cytotoxic metabolite M2 may play a role in the antitumor activity or toxicity of C1311 in the clinic.


Subject(s)
Aminoacridines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Aminoacridines/blood , Aminoacridines/urine , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/urine , Biotransformation , Chromatography, High Pressure Liquid , Feces/chemistry , Female , Glucuronidase/metabolism , Half-Life , Humans , In Vitro Techniques , Male , Mass Spectrometry , Mice , Microsomes, Liver/metabolism
20.
Cancer Res ; 58(23): 5263-6, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9850044

ABSTRACT

The National Cancer Institute uses the hollow fiber assay as part of its screening program for anticancer drug discovery. Angiogenesis to hollow fibers implanted s.c. has not been reported, thereby raising concerns about the efficiency of drug delivery and its subsequent effects on chemosensitivity. By extending postimplantation times beyond the 6-day period presently used, extensive vascular networks develop, resulting in both increased delivery and chemosensitivity to doxorubicin. This study suggests that present protocols used to evaluate compounds may produce false negative results, and additional studies to determine the predictive value of the assay are required.


Subject(s)
Adenocarcinoma/blood supply , Adenocarcinoma/drug therapy , Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Drug Screening Assays, Antitumor/methods , Neovascularization, Pathologic/metabolism , Adenocarcinoma/pathology , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Colonic Neoplasms/pathology , Disease Models, Animal , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Injections, Subcutaneous , Mice , Mice, Nude , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...