Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Crit Rev Oncol Hematol ; 112: 80-102, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28325268

ABSTRACT

In vivo studies in animal models are critical tools necessary to study the fundamental complexity of carcinogenesis. A constant strive to improve animal models in cancer exists, especially those investigating the use of chemotherapeutic effectiveness. In the present systematic review, colorectal cancer (CRC) is used as an example to highlight and critically evaluate the range of reporting strategies used when investigating chemotherapeutic agents in the preclinical setting. A systematic review examining the methodology and reporting of preclinical chemotherapeutic drug studies using CRC murine models was conducted. A total of 45 studies were included in this systematic review. The literature was found to be highly heterogeneous with various cell lines, animal strains, animal ages and chemotherapeutic compounds/regimens tested, proving difficult to compare outcomes between similar studies or indeed gain any significant insight into which chemotherapeutic regimen caused adverse events. From this analysis we propose a minimum core outcome dataset that could be regarded as a standardised way of reporting results from in vivo experimentation.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Evaluation, Preclinical/standards , Animals , Disease Models, Animal , Humans
2.
Br J Cancer ; 102(11): 1555-77, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20502460

ABSTRACT

Animal experiments remain essential to understand the fundamental mechanisms underpinning malignancy and to discover improved methods to prevent, diagnose and treat cancer. Excellent standards of animal care are fully consistent with the conduct of high quality cancer research. Here we provide updated guidelines on the welfare and use of animals in cancer research. All experiments should incorporate the 3Rs: replacement, reduction and refinement. Focusing on animal welfare, we present recommendations on all aspects of cancer research, including: study design, statistics and pilot studies; choice of tumour models (e.g., genetically engineered, orthotopic and metastatic); therapy (including drugs and radiation); imaging (covering techniques, anaesthesia and restraint); humane endpoints (including tumour burden and site); and publication of best practice.


Subject(s)
Animal Experimentation/standards , Animal Welfare/standards , Neoplasms/pathology , Neoplasms/therapy , Practice Guidelines as Topic , Algorithms , Animal Experimentation/ethics , Animal Welfare/ethics , Animal Welfare/organization & administration , Animals , Biomarkers, Pharmacological/analysis , Biomedical Research/ethics , Biomedical Research/legislation & jurisprudence , Biomedical Research/organization & administration , Biomedical Research/standards , Cell Line, Transformed , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Male , Mice , Neoplasm Transplantation/methods , Neoplasm Transplantation/pathology , Neoplasm Transplantation/standards , Neoplasms/diagnosis , Neoplasms/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Br J Radiol ; 81 Spec No 1: S21-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18819995

ABSTRACT

A number of pre-clinical studies have suggested that blocking vascular endothelial growth factor (VEGF) signalling can be beneficial in combination with radiotherapy. This study investigated the effects of cediranib, a highly potent orally available inhibitor of VEGF receptor tyrosine kinase activity in combination with radiation in Calu-6 lung xenografts. In nude mice, Calu-6 tumours were established and treatments initiated at a volume of 250 mm(3). Tumour-localized radiotherapy was given as three or five daily fractions of 2 Gy. Cediranib (3 mg kg(-1)) was administered 2 h prior to each fraction and continued post radiotherapy (concomitant regimen) or was initiated immediately after the completion of radiotherapy (sequential regimen). The endpoint was the time taken for tumour volume to quadruple (RTV4). Combined treatments resulted in a significantly enhanced growth delay compared with either modality alone. The therapeutic benefit was the same irrespective of the scheduling regimen. Tumour regression was observed post radiotherapy, which was associated with high levels of apoptosis and necrosis, and pronounced antivascular effects in histological samples. The amplified antivascular effect of cediranib when given after radiation suggests that pre-irradiated endothelium is sensitized to cediranib. Concomitant 5-day treatment with both cediranib and radiation reduced vessel density, perfusion and increased in tumour hypoxia. This was not associated with an acquired radioresistance suggesting that the maintenance of cediranib treatment post radiotherapy prevents the contribution of hypoxic cells to tumour regrowth. Collectively, these data support the contention that VEGFR inhibition can enhance radiation response in pre-clinical models and provide a rationale to develop cediranib in combination with radiotherapy in the clinical setting.


Subject(s)
Carcinoma/pathology , Lung Neoplasms/pathology , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Apoptosis/drug effects , Carcinoma/drug therapy , Carcinoma/radiotherapy , Combined Modality Therapy , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Mice , Mice, Nude , Necrosis
4.
NMR Biomed ; 21(1): 42-52, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17458919

ABSTRACT

The effect of two novel therapeutic agents on tumour haemodynamics was investigated using a fast dynamic contrast-enhanced (DCE)-MRI protocol (0.5 s/image) sensitive to signal changes in both the vascular input function and tumour during the administration of the macromolecular rapid clearance blood pool agent (MM-RCBPA), gadomelitol (P792, Vistarem). This enabled simultaneous measurement of the tumour blood flow per unit volume of tissue (F/V(T), mL/s/mL), the fractional plasma volume (V(p), %), and the permeability surface area product per unit volume of tissue (PSrho, s(-1)) in subcutaneous SW620 human colorectal tumour xenografts grown in nude rats before and after (at 0 and 22 h; imaging at 24 h) acute treatment with AZD2171 (3 mg/kg) and vandetanib (ZD6474, Zactima; 50 mg/kg), which have inhibitory activity against vascular endothelial growth factor receptor-2 (VEGFR-2) tyrosine kinase. MRI was performed at 4.7 T using a single-slice, modified, T(1)-weighted, spoiled gradient-echo technique. Both compounds reduced gadomelitol uptake into the tumour. AZD2171 and vandetanib, respectively, (a) greatly reduced PSrho to 19.7 +/- 9.5% and 28.9 +/- 14.1% of baseline (P = 0.007 and P = 0.02), (b) markedly reduced V(p) to 31.2 +/- 19.1% and 54.8 +/- 21.2% of baseline (P = 0.015 and P = 0.09), and (c) had no significant effect on F/V(T). There was no significant difference between groups treated with AZD2171 and vandetanib when each variable was compared. The reductions in PSrho and V(p) are consistent with inhibition of VEGF signalling. AZD2171 (3 mg/kg) and vandetanib (50 mg/kg) were also found to produce a comparable chronic inhibition of SW620 tumour growth (89% for both). This study shows that DCE-MRI using an MM-RCPBA can be used to distinguish tumour vascular flow, volume, and permeability surface area product in a tumour model, and enables the acute effects of VEGF signalling inhibition to be examined in detail.


Subject(s)
Colonic Neoplasms/physiopathology , Hemodynamics/drug effects , Heterocyclic Compounds/pharmacology , Magnetic Resonance Imaging/methods , Organometallic Compounds/pharmacology , Piperidines/pharmacology , Quinazolines/pharmacology , Animals , Calibration , Cell Proliferation/drug effects , Colonic Neoplasms/blood supply , Colonic Neoplasms/pathology , Contrast Media , Disease Models, Animal , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Heterocyclic Compounds/metabolism , Humans , Male , Neoplasm Transplantation , Organometallic Compounds/metabolism , Rats , Rats, Nude
5.
Carcinogenesis ; 27(10): 2133-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16782971

ABSTRACT

The Apc(Min/+) mouse model is a clinically relevant model of early intestinal cancer. We used AZD2171, an oral, highly potent and selective vascular endothelial growth factor (VEGF) signaling inhibitor, to investigate the role of VEGF receptor-2 (VEGFR-2) signaling in adenoma development and growth in Apc(Min/+) mice. AZD2171 (5 mg/kg body wt/day) was administered once daily for 28 days to 6-week-old (early-intervention) or 10-week-old (late intervention) mice. In the early-intervention study, AZD2171 reduced the number of macroscopic polyps in the small bowel and colon. Macropolyp diameter was lower in the small bowel, but remained unchanged in the colon. In animals receiving AZD2171, microscopic evaluation of the small intestine showed a significant reduction in the number of larger lesions. In the late-intervention study, AZD2171 treatment reduced macropolyp diameter (but not number) in the small intestine. Microscopic analysis revealed that AZD2171 significantly reduced the number of larger micropolyps in the small bowel, with no large micropolyps present in the colon. AZD2171 treatment had no effect on microvessel density or localization of beta-catenin staining in adenomas or non-tumor intestinal tissue, but significantly reduced the number of cells expressing VEGFR-2 mRNA. In conclusion, the effects of AZD2171 in the small intestine of Apc(Min/+) mice are consistent with an antiangiogenic mechanism of action, limiting growth of adenomas to < or =1 mm. These data also suggest that an early step in adenoma development may depend on VEGFR-2 signaling. Together, these results indicate that VEGFR-2 signaling may play key roles in the development and progression of intestinal adenomas.


Subject(s)
Adenoma/prevention & control , Genes, APC/physiology , Intestinal Neoplasms/prevention & control , Intestinal Polyps/drug therapy , Quinazolines/therapeutic use , Signal Transduction/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Female , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Splenomegaly/prevention & control , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
Br J Cancer ; 92 Suppl 1: S6-13, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15928657

ABSTRACT

Angiogenesis is crucial for maintaining the supply of oxygen and nutrients required to support solid tumour growth. Inhibitors of tumour blood vessel formation are therefore being sought, in particular, inhibitors of vascular endothelial growth factor-A (VEGF)-signalling, which has a pivotal role in stimulating neovascular growth and survival. ZD6474 is an orally bioavailable inhibitor of VEGF receptor-2 tyrosine kinase activity that in preclinical studies has been shown to inhibit both VEGF-induced signalling in endothelial cells and tumour-induced angiogenesis. Consistent with inhibition of angiogenesis, once-daily oral dosing of ZD6474 produced significant broad-spectrum antitumour activity in a panel of histologically diverse human tumour xenografts. In addition to its antiangiogenic properties, ZD6474 also has activity against the epidermal growth factor receptor (EGFR) tyrosine kinase, which could impart a direct inhibitory effect on tumour cell growth and survival. This may be particularly relevant in tumours with a dependency upon EGFR signalling, for example in certain tumours harbouring activating mutations in EGFR. RET kinase has also been identified as a third target for ZD6474. This review summarises preclinical studies with this unique agent and considers its future direction in cancer treatment.


Subject(s)
Neoplasms/physiopathology , Neovascularization, Pathologic , Piperidines/pharmacology , Piperidines/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Administration, Oral , Biological Availability , Cell Survival , Drug Administration Schedule , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Humans , Neoplasms/drug therapy , Neoplasms/enzymology , Piperidines/administration & dosage , Quinazolines/administration & dosage , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/genetics , Signal Transduction
7.
Angiogenesis ; 7(4): 347-54, 2004.
Article in English | MEDLINE | ID: mdl-15886878

ABSTRACT

The proangiogenic vascular endothelial growth factor-A (VEGF) is essential for the development of new tumor vessels. ZD6474 is a novel inhibitor of VEGF receptor-2 (VEGFR-2) tyrosine kinase activity, which also has additional activity against epidermal growth factor (EGF) receptor tyrosine kinase. The antitumor activity of different schedules of ZD6474 in a clinically relevant, metastasizing, murine renal cell carcinoma (RENCA) model was evaluated in this study. RENCA cells were inoculated into the left kidney of 24 mice (day 0). Daily ZD6474 (50 mg/kg p.o.) treatment was initiated 1 day or 10 days after tumor cell inoculation and continued until day 21. Following treatment, kidney weight and volume were assessed and blood vessel density determined by CD31 staining. Visible metastases in the lungs, spleen, and lymph nodes were quantified using a dissection microscope. In an additional study, animals were treated according to the same regimen and quantitative three-dimensional microvascular corrosion casting was performed to enable detailed assessment of the tumor vascular architecture. Therapy initiated on day 1 or day 10 resulted in a 79% and 59% reduction in primary tumor volume, a 79% and 60% reduction in the number of lung metastases, and a 58% and 59% reduction in vessel density of primary tumors compared with the control group, respectively. Corrosion casting proved a 5.4- and 3.2-fold lower vascular volume compared with untreated tumors, observations that paralleled with significant architectural alterations. In this RENCA model, ZD6474 was a highly active inhibitor of tumor angiogenesis, primary tumor growth and tumor metastasis.


Subject(s)
Blood Vessels/drug effects , Carcinoma, Renal Cell/blood supply , Enzyme Inhibitors/pharmacology , Kidney Neoplasms/blood supply , Neovascularization, Pathologic/prevention & control , Piperidines/pharmacology , Quinazolines/pharmacology , Animals , Blood Vessels/ultrastructure , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/ultrastructure , Cell Division/drug effects , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/ultrastructure , Male , Mice , Mice, Inbred BALB C , Microscopy, Electron, Scanning , Neoplasm Metastasis , Neoplasm Transplantation
8.
Magn Reson Imaging ; 21(8): 845-51, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14599534

ABSTRACT

We describe the use of perfusion-permeability magnetic resonance imaging (ppMRI) to study hemodynamic parameters in human prostate tumor xenografts, following treatment with the vascular endothelial growth factor-A (VEGF) receptor tyrosine kinase inhibitor, ZD4190. Using a macromolecular contrast agent (P792), a fast MR imaging protocol and a compartmental data analysis, we were able to demonstrate a significant simultaneous reduction in tumor vascular permeability, tumor vascular volume and tumor blood flow (43%, 30% and 42%, respectively) following ZD4190 treatment (100 mg/kg orally, 24 h and 2 h prior to imaging). This study indicates that MR imaging can be used to measure multiple hemodynamic parameters in tumors, and that tumor vascular permeability, volume and flow, can change in response to acute treatment with a VEGF signaling inhibitor.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Contrast Media , Magnetic Resonance Angiography , Neoplasms, Experimental/blood supply , Quinazolines/therapeutic use , Triazoles/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Capillary Permeability/drug effects , Cell Line, Tumor , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/pathology
9.
Br J Cancer ; 89(10): 1889-95, 2003 Nov 17.
Article in English | MEDLINE | ID: mdl-14612898

ABSTRACT

Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), using gadopentetate dimeglumine, was used to monitor acute effects on tumour vascular permeability following inhibition of vascular endothelial growth factor-A (VEGF-A) signal transduction. Mice bearing PC-3 human prostate adenocarcinoma xenografts were treated with ZD6474, a VEGF receptor-2 (KDR) tyrosine kinase inhibitor. The pharmacokinetic parameter K(trans) was obtained, which reflects vascular permeability and perfusion. Mice were imaged immediately before, and following, acute treatment with ZD6474 (12.5-100 mg x kg(-1) orally). Whole tumours were analysed to obtain mean K(trans) values, and a histogram approach was used to examine intratumour heterogeneity. Reproducibility of K(trans) measurements gave inter- and intra-animal coefficients of variation of 40 and 18%, respectively. Dose-related reductions in K(trans) were evident following acute ZD6474 treatment. A K(trans) reduction of approximately 30% (P<0.001) was evident with 50 and 100 mg x kg(-1) ZD6474, a reduction of 12.5% (P<0.05) at 25 mg x kg(-1), and a reduction that did not reach statistical significance at 12.5 mg kg(-1). A correlation between this dose response and the growth inhibitory effect of ZD6474 following chronic treatment was also observed. The histogram analysis of the data indicated that ZD6474-induced a K(trans) reduction in both the most enhancing rim and the core of PC-3 tumours. Dynamic contrast-enhanced magnetic resonance imaging may have a role in assessing the acute effects of VEGF signalling inhibition, in clinical dose-ranging studies.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Contrast Media/administration & dosage , Gadolinium DTPA/administration & dosage , Magnetic Resonance Imaging/methods , Piperidines/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Quinazolines/therapeutic use , Adenocarcinoma/blood supply , Animals , Capillary Permeability , Dose-Response Relationship, Drug , Humans , Male , Mice , Neoplasms, Experimental , Piperidines/administration & dosage , Piperidines/pharmacology , Prostatic Neoplasms/blood supply , Quinazolines/administration & dosage , Quinazolines/pharmacology , Reproducibility of Results , Signal Transduction , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/antagonists & inhibitors
10.
Microcirculation ; 9(6): 513-22, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12483548

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) increases vascular permeability and angiogenesis in many pathological conditions including cancer, arthritis, and diabetes. VEGF activates VEGF-Receptor 1(VEGF-R1) and VEGF-Receptor 2 (VEGF-R2), which autophosphorylate to initiate a signaling cascade resulting in angiogenesis and increased microvascular permeability. Here we describe a novel VEGF-R2 selective inhibitor, ZM323881 (5-[[7-(benzyloxy) quinazolin-4-yl]amino]-4-fluoro-2-methylphenol), that is a potent and selective inhibitor of VEGF-R2 tyrosine kinase in vitro (IC(50) < 2 nM), compared with other receptor tyrosine kinases, including VEGF-R1 (IC(50) > 50 microM). METHODS: Endothelial cell proliferation was assayed by (3)H-thymidine incorporation in response to VEGF-A +/- ZM323881. The effect of ZM323881 on VEGF-mediated permeability was measured in frog microvessels using the Landis Michel technique. To ensure that ZM323881 was effective in frogs, western analysis was performed on protein extracted from frog lungs incubated in the presence or absence of VEGF-A or VEGF-A with ZM323881. RESULTS: ZM323881 inhibits VEGF-A-induced endothelial cell proliferation (IC(50) = 8 nM) and VEGF-R2 tyrosine phosphorylation in vitro. VEGF-A-mediated increases in vascular permeability in perfused mesenteric microvessels in vivo were reversibly abolished by both ZM323881 and the class III receptor tyrosine kinase inhibitor PTK787/ZK222584. CONCLUSIONS: These data suggest that VEGF-R2 phosphorylation is necessary for VEGF-A-mediated increases in microvascular permeability in vivo.


Subject(s)
Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Capillary Permeability/drug effects , Cell Division/drug effects , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Lung/chemistry , Microcirculation/drug effects , Phosphorylation/drug effects , Proteins/analysis , Ranidae , Signal Transduction , Vascular Endothelial Growth Factor A/physiology
11.
Br J Cancer ; 86(7): 1157-61, 2002 Apr 08.
Article in English | MEDLINE | ID: mdl-11953865

ABSTRACT

The effect of ZD1839 ('Iressa'), a specific inhibitor of the tyrosine kinase activity of the epidermal growth factor receptor, on the radiation response of human tumour cells (LoVo colorectal carcinoma) was evaluated in vitro and in vivo. ZD1839 (0.5 microM, incubated days 1-5) significantly increased the anti-proliferative effect of fractionated radiation treatment (2 Gy day(-1), days 1-3) on LoVo cells grown in vitro (P=0.002). ZD1839 combined with either single or fractionated radiotherapy in mice bearing LoVo tumour xenografts, also produced a highly significant increase in tumour growth inhibition (P< or = 0.001) when compared to treatment with either modality alone. The radio-potentiating effect of ZD1839 was more apparent when radiation was administered in a fractionated protocol. This phenomenon may be attributed to an anti proliferative effect of ZD1839 on tumour cell re-population between radiotherapy fractions. These data suggest radiotherapy with adjuvant ZD1839 could enhance treatment response. Clinical investigation of ZD1839 in combination with radiotherapy is therefore warranted.


Subject(s)
Carcinoma/pathology , Colorectal Neoplasms/pathology , Enzyme Inhibitors/pharmacology , Quinazolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma/radiotherapy , Cell Division , Chemotherapy, Adjuvant , Colorectal Neoplasms/radiotherapy , Gefitinib , Humans , Mice , Transplantation, Heterologous
13.
Cancer Res ; 60(4): 970-5, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10706112

ABSTRACT

There is evidence that vascular endothelial growth factor (VEGF) contributes to solid tumor growth through the promotion of both angiogenesis and tumor vascular permeability. To abrogate VEGF signaling, we developed a small molecular weight inhibitor of VEGF receptor tyrosine kinase (RTK) activity that was compatible with chronic oral administration. ZD4190, a substituted 4-anilinoquinazoline, is a potent inhibitor of KDR and Flt-1 RTK activity, and VEGF stimulated HUVEC proliferation in vitro. Chronic once-daily oral dosing of ZD4190 to young rats produced a dose-dependent increase in the femoral epiphyseal growth plate area, which may be attributed to the inhibition of VEGF signaling in vivo because vascular invasion of cartilage is a prerequisite to the process of ossification. Once-daily oral dosing of ZD4190 to mice bearing established (approximately 0.5 cm3) human tumor xenografts (breast, lung, prostate, and ovarian) elicited significant antitumor activity and at doses that would not be expected to have any direct antiproliferative effect on tumor cells. Prolonged tumor cytostasis was further demonstrated in a PC-3 xenograft model with 10 weeks of ZD4190 dosing, and upon withdrawal of therapy, tumor growth resumed after a short delay. These observations are entirely consistent with the proposed mode of action. ZD4190 is one of a series of VEGF RTK inhibitors that may have utility in the treatment of a range of histologically diverse solid tumor types.


Subject(s)
Antineoplastic Agents/pharmacology , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Triazoles/pharmacology , Administration, Oral , Animals , Epiphyses/drug effects , Epiphyses/pathology , Female , Humans , Hypertrophy , Mice , Neoplasm Transplantation , Rats , Receptors, Vascular Endothelial Growth Factor , Transplantation, Heterologous , Tumor Cells, Cultured
14.
J Med Chem ; 42(26): 5369-89, 1999 Dec 30.
Article in English | MEDLINE | ID: mdl-10639280

ABSTRACT

A series of substituted 4-anilinoquinazolines and related compounds were synthesized as potential inhibitors of vascular endothelial growth factor (VEGF) receptor (Flt and KDR) tyrosine kinase activity. Enzyme screening indicated that a narrow structure-activity relationship (SAR) existed for the bicyclic ring system, with quinazolines, quinolines, and cinnolines having activity and with quinazolines and quinolines generally being preferred. Substitution of the aniline was investigated and clearly indicated that small lipophilic substituents such as halogens or methyl were preferred at the C-4' position. Small substituents such as hydrogen and fluorine are preferred at the C-2' position. Introduction of a hydroxyl group at the meta position of the aniline produced the most potent inhibitors of Flt and KDR tyrosine kinases activity with IC(50) values in the nanomolar range (e.g. 10, 12, 13, 16, and 18). Investigation of the quinazoline C-6 and C-7 positions indicates that a large range of substituents are tolerated at C-7, whereas variation at the C-6 is more restricted. At C-7, neutral, basic, and heteroaromatic side chains led to very potent compounds, as illustrated by the methoxyethoxy derivative 13 (IC(50) < 2 nM). Our inhibitors proved to be very selective inhibitors of Flt and KDR tyrosine kinase activity when compared to that associated with the FGF receptor (50- to 3800-fold). Observed enzyme profiles translated well with respect to potency and selectivity for inhibition of growth factor stimulated proliferation of human umbilical vein endothelial cells (HUVECs). Oral administration of selected compounds to mice produced total plasma levels 6 h after dosing of between 3 and 49 microM. In vivo efficacy was demonstrated in a rat uterine oedema assay where significant activity was achieved at 60 mg/kg with the meta hydroxy anilinoquinazoline 10. Inhibition of growth of human tumors in athymic mice has also been demonstrated: compound 34 inhibited the growth of established Calu-6 lung carcinoma xenograft by 75% (P < 0.001, one tailed t-test) following daily oral administration of 100 mg/kg for 21 days.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Animals , Biological Availability , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Enzyme Inhibitors/blood , Female , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Organ Size , Quinazolines/blood , Rats , Receptors, Vascular Endothelial Growth Factor , Structure-Activity Relationship , Tumor Cells, Cultured , Uterus/drug effects
15.
Br J Cancer ; 78(9): 1199-202, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9820180

ABSTRACT

Resistance of tumour cells to methylating and monochloroethylating agents in vitro and in vivo has been linked to levels of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). In a clinical trial of temozolomide in advanced malignant melanoma, the relationship between pretreatment MGMT levels in biopsies of cutaneous tumours and involved lymph nodes and clinical response to the drug has been studied. Among 50 evaluable patients, there were three complete responses (CR), four partial responses (PR), six with stable disease (SD) and 37 with progressive disease (PD), with an overall response rate of 14%. In 33 patients in whom MGMT level and clinical response could be evaluated, the tumour MGMT levels (fmol mg(-1) protein) were: CR, 158 +/- 119; PR, 607 +/- 481; NC, 171 +/- 101; PD, 185 +/- 42.3. Thus, measurements of pretreatment levels of MGMT in melanoma did not predict for response to temozolomide.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Dacarbazine/analogs & derivatives , Melanoma/drug therapy , Melanoma/enzymology , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Adult , Biopsy , Dacarbazine/therapeutic use , Female , Humans , Leukocytes, Mononuclear/enzymology , Lymphatic Metastasis , Male , Melanoma/pathology , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/blood , Predictive Value of Tests , Prospective Studies , Temozolomide
16.
Cancer Res ; 58(19): 4363-7, 1998 Oct 01.
Article in English | MEDLINE | ID: mdl-9766665

ABSTRACT

Temozolomide, a methylating imidazotetrazinone, has antitumor activity against gliomas, malignant melanoma, and mycosis fungoides and is presently administered as a 5-day oral schedule every 4 weeks. This Phase I study aimed to determine the maximum tolerated dose of temozolomide administered as a single oral daily dose for a continuous 6- or 7-week period, evaluate the plasma pharmacokinetics on this schedule, and compare total plasma exposure over 7 weeks with the conventional 5-day regimen. Twenty-four patients with varying tumor types (17 of 24 gliomas) received temozolomide. All had clinically evaluable, refractory disease; normal renal, hepatic, and bone marrow function; and WHO performance status < or = 2. Temozolomide was administered at 50 mg/m2/day, increasing by 25 mg/m2/day/cohort until at 100 mg/m2/day grade 4 myelotoxicity forced dose reductions to 85 mg/m2/day, then 75 mg/m2/day. At 75 mg/m2/day the regimen was extended to 7 weeks, allowing the future potential combination with irradiation for primary gliomas. Patient responses (standard Union International Contre Cancer criteria; for gliomas objective response) and toxicity were assessed. Temozolomide plasma pharmacokinetics were determined on day 1 and at the beginning of the final week of administration (n = 5). The most frequent toxicities were myelosuppression and grades 1 and 2 nausea and vomiting. Grade 4 leucopenia and thrombocytopenia occurred in one of four patients receiving 100 mg/m2/day temozolomide and in one of seven patients receiving 85 mg/m2/day. These hematological toxicities did not exceed grade 2 in 10 patients receiving 75 mg/m2/day temozolomide. One of 4 malignant melanoma patients and 7 of 17 glioma patients (41%) demonstrated tumor responses. The overall response rate for this prolonged schedule was 33% (objective response, 7 of 24 patients; partial response, 1 of 24 patients); also, 6 of 17 glioma patients maintained SD. Peak plasma temozolomide concentrations were obtained 30-90 min after oral administration. Elimination in plasma was best described by a monoexponential equation with an elimination half-life of 96 +/- 16 min. No plasma accumulation of temozolomide occurred. Toxicity was greatest in higher dose cohorts, with a resultant maximum tolerated dose of 85 mg/m2/day, whereas lower dose cohorts tolerated the schedule well. The area under the temozolomide plasma versus time curve was noncumulative between the first and last week of the schedule. Temozolomide administration of 75 mg/m2/day over a 7-week period permits a 2.1-fold greater drug exposure/4 weeks in comparison with the 5-day schedule of 200 mg/m2/day repeated every 28 days. The overall response rate was 33% (glioma patients, 41% and a further 25% SD). Temozolomide (75 mg/m2/day) for 7 weeks is the recommended starting dose for further assessment of this schedule.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Neoplasms/drug therapy , Administration, Oral , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/blood , Brain Neoplasms/pathology , Calibration , Chromatography, High Pressure Liquid , Dacarbazine/administration & dosage , Dacarbazine/adverse effects , Dacarbazine/blood , Drug Administration Schedule , England , Female , Glioma/pathology , Humans , Male , Metabolic Clearance Rate , Middle Aged , Neoplasms/pathology , Temozolomide
18.
Anticancer Drugs ; 8(1): 92-7, 1997 Jan.
Article in English | MEDLINE | ID: mdl-9147618

ABSTRACT

The in vitro cytotoxicity of 8-carbamoyl-3-methylimidazo [5,1-d]-1,2,3,5-tetrazine-4(3H)-one (temozolomide) with concurrent X-irradiation was examined in a human glioblastoma cell line (U373MG) as a potential radio-chemotherapeutic treatment for malignant glioma. The combination was also examined in a human colorectal adenocarcinoma (Mawi) which had 100-fold greater O6-alkylguanine-DNA alkyltransferase (AGT) activity, a DNA-repair protein which confers resistance to temozolomide. A comparison of IC50 values indicated U373MG to be over 32-fold more sensitive to temozolomide than Mawi, but slightly more resistant to X-irradiation (p < 0.035; unpaired two-tailed t-test). Temozolomide and X-irradiation proved largely additive in U373MG by isobologram analysis (50% iso-effect) and the addition of 10 microM temozolomide to 1-2 Gy of X-irradiation increased cell kill by 2.5- to 3.0-fold. However, the combination was antagonistic in Mawi: an effect attributed to AGT induction by X-irradiation as the antagonism was removed by co-incubation with the AGT inhibitor O6-benzylguanine (O6-BG 1 microM; 24 h). O6-BG did not affect the radiation dose-response curve, but significantly increased temozolomide cytotoxicity (p < 0.015). In conclusion, the combination of temozolomide with radiation is at best additive, but could nonetheless by of considerable therapeutic benefit in glioma, particularly if administered for prolonged periods. If AGT induction compromises the efficacy of this therapy, it may be circumvented with an appropriate inhibitor such as O6-BG.


Subject(s)
Adenocarcinoma/therapy , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Colonic Neoplasms/therapy , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Combined Modality Therapy , DNA Adducts/drug effects , DNA Adducts/metabolism , DNA Repair/drug effects , Dacarbazine/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Methyltransferases/antagonists & inhibitors , Methyltransferases/metabolism , O(6)-Methylguanine-DNA Methyltransferase , Temozolomide , Tumor Cells, Cultured
19.
Cancer Chemother Pharmacol ; 40(3): 266-72, 1997.
Article in English | MEDLINE | ID: mdl-9219512

ABSTRACT

The purpose of the present study was to examine the effect of O6-benzylguanine (O6-BG) on the antitumour activity and toxicity of 8-carbamoyl-3-methylimidazo [5, 1-d]-1,2,3,5-tetrazine-4(3H)-one (temozolomide) in a human malignant melanoma xenograft model following single and multiple administration of the combination. O6-BG irreversibly inactivates the DNA-repair protein O6-alkylguanine-DNA alkyltransferase (AGT), which confers resistance to temozolomide. Preadministration of O6-BG (35 mg/kg, i.p.) 1 h prior to temozolomide (i.p.) was examined using single and daily x5 dosing regimens in athymic mice bearing subcutaneous A375P xenografts. The AGT activity of A375P tumors was 95 +/- 8 fmol/mg protein (mean +/- SE, n = 4). O6-BG alone completely suppressed xenograft AGT activity within 1 h of administration but had no effect upon tumor growth. O6-BG did not significantly increase the tumor growth delay induced by a single 200-mg/ kg dose of temozolomide (P > 0.05, two-tailed Mann-Whitney test) but did increase the associated mean body weight loss (P < 0.025). In contrast, when the same dose of temozolomide was divided into five equal fractions (40 mg/kg) and given with O6-BG on 5 consecutive days, a comparable increase in toxicity was accompanied by a very significant increase in tumor growth delay (P < 0.0025), equivalent to that produced by a 3-fold greater dose of temozolomide alone. O6-BG with temozolomide also produced a greater antitumour effect than an equitoxic dose of temozolomide alone on this schedule (P < 0.005). These data indicate that the enhancement of temozolomide antitumour activity by O6-BG preadministration is dependent upon the schedule of drug administration, with multiple dosing of O6-BG + temozolomide producing the greatest effect. The results also suggest that prolonged administration of the combination can lead to an increase in the therapeutic index of temozolomide.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma, Experimental/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/toxicity , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/toxicity , Drug Administration Schedule , Guanine/administration & dosage , Guanine/analogs & derivatives , Guanine/toxicity , Humans , Melanoma, Experimental/metabolism , Methyltransferases/drug effects , Methyltransferases/metabolism , Mice , Mice, Nude , Neoplasm Transplantation , O(6)-Methylguanine-DNA Methyltransferase , Temozolomide , Tumor Cells, Cultured
20.
Br J Cancer ; 74(7): 1030-6, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8855970

ABSTRACT

O6-benzylguanine (O6-BG) and 3-aminobenzamide (3-AB) inhibit the DNA repair proteins O6-alkylguanine-DNA alkyltransferase (AGT) and poly(ADP-ribose) polymerase (PARP) respectively. The effect of O6-BG and/or 3-AB on temozolomide and 1,3-bis(2-chloroethyl)-nitrosourea (BCNU) cytotoxicity, was assessed in seven human tumour cell lines: six with an AGT activity of > 80 fmol mg-1 protein (Mer+) and one with an AGT activity of < 3 fmol mg-1 protein (Mer-). Three of the Mer+ cell lines (LS174T, DLD1 and HCT116) were considered to exhibit resistance to methylation by a mismatch repair deficiency (MMR-), each being known to exhibit microsatellite instability, and DLD1 and HCT116 having well-characterised defects in DNA mismatch binding. Potentiation was defined as the ratio between an IC50 achieved without and with a particular inhibitor treatment. Temozolomide or BCNU cytotoxicity was not potentiated by either inhibitor in the Mer- cell line. Preincubation with O6-BG (100 microM for 1 h) was found to potentiate the cytotoxicity of temozolomide by 1.35- to 1.57-old in Mer+/MMR+ cells, but had no significant effect in Mer+/MMR- cells. In comparison, O6-BG pretreatment enhanced BCNU cytotoxicity by 1.94- to 2.57-fold in all Mer+ cell lines. Post-incubation with 3-AB (2 mM, 48 h) potentiated temozolomide by 1.35- to 1.59-fold in Mer+/MMR+ cells, and when combined with O6-BG pretreatment produced an effect which was at least additive, enhancing cytotoxicity by 1.97- to 2.16-fold. 3-AB treatment also produced marked potentiation (2.20- to 3.12-fold) of temozolomide cytotoxicity in Mer+/MMR- cells. In contrast, 3-AB produced marginal potentiation of BCNU cytotoxicity in only three cell lines (1.19- to 1.35-fold), and did not enhance the cytotoxicity of BCNU with O6-BG treatment in any cell line. These data suggest that the combination of an AGT and PARP inhibitor may have a therapeutic role in potentiating temozolomide activity, but that the inhibition of poly(ADP-ribosyl)ation has little effect on the cytotoxicity of BCNU.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , DNA Repair , Neoplasms/drug therapy , Benzamides/administration & dosage , Benzamides/pharmacology , Carmustine/administration & dosage , Carmustine/pharmacology , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Guanine/administration & dosage , Guanine/analogs & derivatives , Guanine/pharmacology , Humans , Neoplasms/enzymology , Neoplasms/genetics , Temozolomide , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...