Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Int J Oncol ; 31(2): 353-60, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17611692

ABSTRACT

Despite promising early data, the natural product dolastatin 10 has not been successful as a single agent in phase II clinical trials. Herein the mechanism of action and efficacy of a synthetic analogue, auristatin PYE, was investigated in 2 human colon adenocarcinoma models, DLD-1 and COLO 205. In vivo efficacy was assessed in subcutaneous xenografts following intravenous administration. Mechanistic studies investigated effects of auristatin PYE on microtubule disruption using immunocytochemistry, whilst cell cycle effects were studied using flow cytometry. Possible effects on tumour functional blood vasculature were assessed in tumour-bearing mice. Auristatin PYE was less potent in vitro than dolastatin 10, but was significantly more effective (p<0.01) in vivo against both tumours. Significant effects on tumour blood vasculature were seen, with optimal shutdown at 6-h post-treatment. Extensive necrosis became more evident over time after treatment. Auristatin PYE caused severe disruption of normal microtubule structure at concentrations and times comparable with the IC50 data, and also instigated a G2/M cell cycle block. Auristatin PYE was more effective in the DLD-1 and COLO 205 models than dolastatin 10, with anti-tumour effects mediated through vascular shutdown. These data suggest that auristatin PYE has good potential as an anti-cancer agent.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Depsipeptides/chemistry , Depsipeptides/pharmacology , Animals , Cell Cycle , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Inhibitory Concentration 50 , Mice , Mice, Nude , Models, Chemical , Necrosis , Neoplasm Transplantation
2.
Int J Oncol ; 29(6): 1493-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17088988

ABSTRACT

We describe a modified hollow fibre assay (HFA) for investigating the potential of novel molecules as pharmaceutical agents. In particular the assay provides drug/target interaction data that can facilitate the selection of lead compounds for further evaluation in more sophisticated solid tumour models, whilst successfully implementing the 3Rs - the 'replacement' 'refinement' and 'reduction' of animals. This more ethical and rapid approach to early drug development does not compromise on the validity, sensitivity, predictivity or efficacy of preclinical evaluation. We present novel data using the standard cross-linker mitomycin C (MMC) as a positive control, and two investigational DNA interactive molecules (C1311/ SJG-136). Tumour cells were seeded in fibres and implanted into mice. Following treatment with an intraperitoneal injection, fibres were excised and cells retrieved for pharmacodynamic analysis using the comet assay/fluorescence microscopy. Microscopy results revealed nuclear uptake and localisation within cytoplasmic organelles of HT29 colorectal adenocarcinoma cells following treatment with C1311 (150 mg/kg). Following treatment with SJG-136 (0.3 mg/kg) a 27.3% (p<0.001) DNA cross-linking (s.c.) effect was observed in the HL60 acute promyelocytic leukaemia cell line. DNA cross-linking effects of 55% (i.p) and 50% (s.c.) (p<0.005) were observed in the A549 lung carcinoma cell line following administration of MMC (6 mg/kg). These data are consistent with previous activity defined using solid tumour models, and support the use of the HFA for in vivo pharmacodynamic investigation whilst significantly reducing animal numbers and the influence of tumour growth on the welfare of mice.


Subject(s)
Aminoacridines/pharmacology , Animal Welfare , Benzodiazepinones/pharmacology , Drug Screening Assays, Antitumor/methods , Mitomycin/pharmacology , Pyrroles/pharmacology , Animals , Cell Growth Processes/physiology , Comet Assay , Drug Screening Assays, Antitumor/ethics , Female , HL-60 Cells , HT29 Cells , Humans , Mice , Microscopy, Fluorescence/methods , Neoplasm Transplantation , Transplantation, Heterologous
3.
Clin Cancer Res ; 11(3): 971-81, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15709162

ABSTRACT

The number of anticancer agents that fail in the clinic far outweighs those considered effective, suggesting that the selection procedure for progression of molecules into the clinic requires improvement. The value of any preclinical model will ultimately depend on its ability to accurately predict clinical response. This review focuses on the major contributions of preclinical screening models to anticancer drug development over the past 50 years. Over time, a general transition has been observed from the empirical drug screening of cytotoxic agents against uncharacterized tumor models to the target-orientated drug screening of agents with defined mechanisms of action. New approaches to anticancer drug development involve the molecular characterization of models along with an appreciation of the pharmacodynamic and pharmacokinetic properties of compounds [e.g., the US National Cancer Institute (NCI) in vitro 60-cell line panel, hollow fiber assay, and s.c. xenograft]. Contributions of other potentially more clinically relevant in vivo tumor models including orthotopic, metastatic, and genetically engineered mouse models are also reviewed. Although this review concentrates on the preclinical screening efforts of the NCI, European efforts are not overlooked. Europe has played a key role in the development of new anticancer agents. The two largest academic drug development groups, the European Organisation for Research and Treatment of Cancer and Cancer Research UK, have been collaborating with the NCI in the acquisition and screening of compounds since the 1970s. As with the drug development process internationally, rational pharmacodynamic approaches have more recently been adopted by these two groups.


Subject(s)
Drug Screening Assays, Antitumor/methods , Neoplasms/drug therapy , Animals , Disease Models, Animal , Drug Screening Assays, Antitumor/trends , Humans
4.
Clin Cancer Res ; 11(2 Pt 1): 416-20, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15701823

ABSTRACT

The therapeutic potential of targeting the tumor vascular supply is now widely recognized. Intense research and development activity has resulted in a variety of investigational agents, a number of which are currently in clinical development. As these novel agents are quite distinct from the cytotoxic drugs conventionally used in the treatment of solid tumors, it will be particularly important to ensure early differentiation of these vascular-targeted therapies in order to encourage widespread understanding of their potential benefits and application in the clinic. Two distinct groups of vascular-targeted therapies have evolved: antiangiogenic agents and vascular-disrupting approaches. These differ in three key respects: their physiologic target, the type or extent of disease that is likely to be susceptible, and the treatment scheduling. Inhibitors of angiogenesis interfere with new vessel formation and therefore have a preventative action, require chronic administration, and are likely to be of particular benefit in early-stage or asymptomatic metastatic disease. Vascular-disrupting agents target the established tumor blood vessels, resulting in tumor ischemia and necrosis. These agents are therefore given acutely, show more immediate effects, and may have particular efficacy against advanced disease. It is essential that these agents can be readily distinguished from conventional therapies and that an understanding of key differences between the two types of vascular-targeted therapies is fostered. Here, a simple taxonomy and nomenclature is proposed in anticipation that the therapeutic potential of this novel class can be realized as these approaches advance in clinical settings and a new anticancer strategy becomes available in the clinic.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Cell Differentiation , Neoplasms/blood supply , Neoplasms/therapy , Neovascularization, Pathologic/therapy , Animals , Growth Substances/physiology , Humans , Neovascularization, Pathologic/physiopathology
5.
Clin Cancer Res ; 11(22): 8172-9, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16299249

ABSTRACT

BACKGROUND: In previous experiments, we showed that heparin oligosaccharides inhibit the angiogenic cytokine fibroblast growth factor-2. Here, we present the first in vivo study of size-fractionated heparin oligosaccharides in four models of angiogenesis that are progressively less dependent on fibroblast growth factor-2. EXPERIMENTAL DESIGN: Heparin oligosaccharides were prepared using size-exclusion gel filtration chromatography and characterized through depolymerization and strong anion exchange high-performance liquid chromatography. Size-defined oligosaccharides (20 mg/kg/d) were given to mice bearing s.c. sponges that were injected with fibroblast growth factor-2 (100 ng/d). After 14 days, octasaccharides and decasaccharides reduced the microvessel density to levels below control. In a second experiment, HEC-FGF2 human endometrial cancer cells that overexpress fibroblast growth factor-2 were implanted in a hollow fiber placed s.c. in vivo. Oligosaccharides were given at 20 mg/kg/d for 2 weeks and the data again showed that octasaccharides significantly reduced microvessel density around the fiber (P = 0.03). In a more complex model, where angiogenesis was induced by a broad spectrum of growth factors, including vascular endothelial growth factor, we implanted H460 lung carcinoma cells in hollow fibers and treated the animals with oligosaccharides at 20 mg/kg/d over 3 weeks. Octasaccharides reduced the microvessel density to that of control. Preliminary investigation of 6-O-desulfated heparins showed that these also had antiangiogenic activity. RESULTS: Finally, we examined the inhibitory potential of hexasaccharides and octasaccharides given at 20 mg/kg/d and these inhibited the growth of H460 lung carcinoma in vivo. At clinically attainable concentrations, significant anticoagulation (activated partial thromboplastin time, anti-factor Xa, and anti-factor IIa) was not observed in vitro unless species containing > or =16 saccharide residues were investigated. CONCLUSIONS: Thus, our preclinical data show that heparin octasaccharides represent novel antiangiogenic compounds that can be given without the anticoagulant effects of low molecular weight heparin.


Subject(s)
Lung Neoplasms/prevention & control , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Oligosaccharides/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Anticoagulants/pharmacology , Cell Line, Tumor , Female , Fibroblast Growth Factor 2/pharmacology , Heparin/chemistry , Heparin/pharmacology , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Partial Thromboplastin Time , Xenograft Model Antitumor Assays/instrumentation , Xenograft Model Antitumor Assays/methods
6.
Eur J Med Chem ; 41(3): 330-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16458393

ABSTRACT

The protein O6-alkylguanine-DNA alkyltransferase (Atase) is responsible for the repair of DNA lesions generated by several clinically important anti-cancer drugs; this is manifest as active resistance in those cancer cell lines proficient in Atase expression. Novel O6-substituted guanine analogues have been synthesized, bearing acidic, basic and hydrogen bonding functional groups. In contrast to existing O6-modified purine analogues, such as methyl or benzyl, the new compounds were found to resist repair by Atase even when tested at concentrations much higher than O6-benzylguanine, a well-established Atase substrate active both in vitro and in vivo. The inactivity of the new purines as covalent substrates for Atase indicates that agents to deliver these groups to DNA would represent a new class of DNA-modifying drug that circumvents Atase-mediated resistance.


Subject(s)
DNA Repair Enzymes/chemistry , Drug Design , Guanine/analogs & derivatives , Guanine/chemistry , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Purines/chemistry , Crystallography, X-Ray , DNA/chemistry , DNA/drug effects , Guanine/pharmacology , Humans , Models, Biological , Molecular Structure , Mutation , O(6)-Methylguanine-DNA Methyltransferase/genetics , Purines/pharmacology , Substrate Specificity
7.
Clin Cancer Res ; 10(19): 6677-85, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15475458

ABSTRACT

PURPOSE: The hollow fiber assay is used successfully as a routine in vivo screening model to quantitatively define anticancer activity by the National Cancer Institute. This study investigates whether the hollow fiber assay can be used as a short-term in vivo model to demonstrate specific pharmacodynamic end points, namely microtubule and cell cycle disruption. EXPERIMENTAL DESIGN: The growth of A549 cells was characterized within hollow fibers over 5 days in vivo at both subcutaneous (s.c.) and intraperitoneal (i.p.) sites. Drugs were administered on day 4 (i.p.). RESULTS: At 24 hours, cells were retrieved from fibers at both i.p. and s.c. sites of paclitaxel-treated (20 mg/kg) and combretastatin A1 phosphate-treated (150 mg/kg) mice. Cell cycle analysis after paclitaxel treatment revealed a mean G(2)-M phase population of 48.04% (i.p.) and 25.76% (s.c.) compared with vehicle group mice (6.78 and 5.56%, respectively; P = <0.001 and 0.005, respectively). Tumor cells retrieved from combretastatin A1 phosphate-treated mice had a mean G2-M phase population of 36.3% (i.p.) and 29.36% (s.c.) compared with cells retrieved from vehicle group mice (5.58 and 5.49%, respectively; P = <0.001). Using fluorescence and laser-confocal microscopy, paclitaxel was revealed to induce the formation of spindle asters and tubulin polymerization. Combretastatin A1 phosphate was shown to hold cells in mitosis. Changes in nuclear morphology were also observed. CONCLUSION: These data demonstrate that the hollow fiber assay can be used as a short-term in vivo model for studying the pharmacodynamic effects of both standard and novel compounds on microtubules. Evidence has also been provided to support the routine use of the in vivo hollow fiber assay for demonstrating the mechanism of action of a drug.


Subject(s)
Drug Screening Assays, Antitumor/methods , Microtubules/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor/instrumentation , Female , Flow Cytometry , Humans , Immunohistochemistry , Membranes, Artificial , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Fluorescence , Microtubules/drug effects , Neoplasm Transplantation/methods , Paclitaxel/pharmacology , Polyvinyls , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Stilbenes/pharmacology , Transplantation, Heterologous , Tubulin/analysis , Tubulin/metabolism
8.
Mol Cancer Ther ; 3(12): 1565-75, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15634650

ABSTRACT

Phortress is a novel, potent, and selective experimental antitumor agent. Its mechanism of action involves induction of CYP1A1-catalyzed biotransformation of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (5F 203) to generate electrophilic species, which covalently bind to DNA, exacting lethal damage to sensitive tumor cells, in vitro and in vivo. Herein, we investigate the effects of DNA adduct formation on cellular DNA integrity and progression through cell cycle and examine whether a relevant pharmacodynamic end point may be exploited to probe the clinical mechanism of action of Phortress and predict tumor response. Single cell gel electrophoresis (SCGE) was applied to quantify DNA damage and cell cycle analyses conducted upon 5F 203 treatment of benzothiazole-sensitive MCF-7 and inherently resistant MDA-MB-435 breast carcinoma cells. Following treatment of xenograft-bearing mice and mice possessing hollow fiber implants containing MCF-7 or MDA-MB-435 cells with Phortress (20 mg/kg, i.p., 24 hours), tumor cells and xenografts were recovered for analyses by SCGE. Dose- and time-dependent DNA single and double strand breaks occurred exclusively in sensitive cells following treatment with 5F 203 in vitro (10 nmol/L-10 micromol/L; 24-72 hours). In vivo, Phortress-sensitive and Phortress-resistant tumor cells were distinct; moreover, DNA damage in xenografts, following treatment of mice with Phortress, could be determined. Interrogation of the mechanism of action of 5F 203 in silico by self-organizing map-based cluster analyses revealed modulation of phosphatases and kinases associated with cell cycle regulation, corroborating observations of selective cell cycle perturbation by 5F 203 in sensitive cells. By conducting SCGE, tumor sensitivity to Phortress, an agent currently undergoing clinical evaluation, may be determined.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , DNA Adducts/drug effects , Drug Resistance, Neoplasm , Thiazoles/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Cycle/drug effects , Comet Assay , Computational Biology , Cytochrome P-450 CYP1A1/metabolism , DNA Damage/drug effects , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Membranes, Artificial , Mice , Mice, Nude , Time Factors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 3(11): 1451-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542784

ABSTRACT

The use of many common clinically relevant chemotherapeutics is often limited due to insufficient delivery to the tumor and dose-limiting systemic toxicities. Therefore, therapeutics that specifically target tumor cells and are nontoxic to normal cells are required. Here, we report the development of a novel class of liposomes composed of lipid prodrugs, which use the increased secretory phospholipase A2 type IIA (sPLA2) activity of the tumor microenvironment as a trigger for the release of anticancer etherlipids (AEL). Treatment of sPLA2-secreting tumor cells in vitro with liposomes consisting of proAELs resulted in growth inhibition comparable with addition of the AELs alone. Using a specific sPLA2 inhibitor, we showed the low cytotoxicity of the nonhydrolyzed proAEL liposomes and have proven the sPLA2 dependency of the activation of proAELs to cytotoxic AELs. In addition, we showed that our proAEL liposomes circumvent the inherent hemolytic toxicities associated with the use of etherlipids, thereby allowing i.v. administration of such therapeutics as nontoxic prodrug liposomes. Furthermore, using a sPLA2-secreting human colon cancer xenograft model, we showed that the proAEL liposomes are capable of inducing a tumor growth delay in vivo. Taken together, these data support the validity of this novel tumor-selective liposomal prodrug delivery strategy. This new approach also provides a promising system for tumor-selective delivery and release of conventional chemotherapeutics encapsulated in the sPLA2-degradable prodrug liposomes.


Subject(s)
Antineoplastic Agents/pharmacology , Lipids/pharmacology , Liposomes/administration & dosage , Neoplasms/metabolism , Phospholipases A/metabolism , Prodrugs/administration & dosage , Prodrugs/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Catalysis/drug effects , Cell Line, Tumor , Drug Delivery Systems , Ether/administration & dosage , Ether/chemistry , Ether/pharmacology , Ether/toxicity , Female , Hemolysis/drug effects , Humans , Hydrolysis/drug effects , Lipids/administration & dosage , Lipids/chemistry , Lipids/toxicity , Mice , Molecular Structure , Neoplasms/enzymology , Neoplasms/pathology , Organ Specificity , Phospholipases A/antagonists & inhibitors , Phospholipases A2 , Prodrugs/chemistry , Prodrugs/toxicity , Xenograft Model Antitumor Assays
10.
Mol Cancer Ther ; 1(4): 239-46, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12467219

ABSTRACT

Novel 2-(4-aminophenyl)benzothiazoles (e.g., compounds 1 and 2) possess highly selective, potent antitumor properties in vitro and in vivo. Elucidation of the mechanism of action of this structurally simple class of compounds has occurred in parallel with selection of a candidate clinical agent. Antitumor benzothiazoles induce and are biotransformed by cytochrome P 450 1A1 to putative active, as well as inactive metabolites. Metabolic inactivation of the molecule has been thwarted by isosteric replacement of hydrogen with fluorine atoms at positions around the benzothiazole nucleus. Amino acid conjugation to the exocyclic primary amine function of 2-(4-aminophenyl)benzothiazoles has been used to overcome limitations posed by drug lipophilicity. Water soluble, chemically stable prodrugs rapidly and quantitatively revert to their parent amine in mice, rats, and dogs in vivo. Plasma concentrations of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole (2) regenerated from the lysylamide prodrug (2b), sufficient to elicit cytocidal activity against ZR-75-1 and T47D human mammary carcinoma cell lines persist > 6 h. The growth of breast (MCF-7) and ovarian (IGROV-1) xenograft tumors is significantly retarded by 2b. Manageable toxic side effects are reported from preclinically efficacious doses of 2b. Cytochrome P 450 1A1 protein expression, selectively induced in sensitive carcinoma cells, was detected in MCF-7 and IGROV-1 tumors 24 h after treatment of mice with 2b (20 mg/kg). The lysyl amide prodrug of 2-(4-amino-3-methylphenyl)-5-fluorobenzothiazole is potentially suitable for clinical evaluation.


Subject(s)
Amino Acids/metabolism , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Ovarian Neoplasms/drug therapy , Thiazoles/therapeutic use , Aniline Compounds/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Benzothiazoles , Blotting, Western , Cell Division/drug effects , Cytochrome P-450 CYP1A1/metabolism , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Female , Humans , Male , Mammary Neoplasms, Experimental/enzymology , Mice , Mice, Nude , Ovarian Neoplasms/enzymology , Prodrugs , Rats , Rats, Sprague-Dawley , Thiazoles/pharmacokinetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
11.
Mol Cancer Ther ; 2(6): 527-34, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12813131

ABSTRACT

The cytochrome P450 family of enzymes is involved in the Phase I metabolism of a wide variety of compounds. Although generally involved with detoxification, overexpression of one family member, cytochrome P450 1B1 (CYP1B1), has been associated with human epithelial tumors. As such, CYP1B1 was hypothesized to be a novel target for the development of anticancer therapies. We investigated expression of CYP1B1 protein in 61 human colorectal adenocarcinomas and compared this to that observed in 14 histologically normal human large bowel samples removed from patients undergoing surgery for large bowel tumors. Although we confirmed that CYP1B1 was expressed at high levels in human colorectal tumor epithelia, we also found that CYP1B1 was not absent from normal colonic epithelia but was expressed at low levels. The expression of CYP1B1 in colon tumors does not correlate with tumor stage or degree of lymph node invasion in this study. Furthermore, in addition to expression in colon epithelia, CYP1B1 is also observed in blood vessels within the colon. As with the epithelia, levels of CYP1B1 were higher in tumor vasculature than that of the normal colon. Although these observations greatly support the development of CYP1B1 targeted anticancer therapies, they also indicate the caution that should be observed when developing such drugs.


Subject(s)
Adenocarcinoma/enzymology , Aryl Hydrocarbon Hydroxylases/biosynthesis , Colonic Neoplasms/enzymology , Adenocarcinoma/pathology , Blotting, Western , Cell Line, Tumor , Colonic Neoplasms/pathology , Colorectal Neoplasms/pathology , Cytochrome P-450 CYP1B1 , Epithelial Cells/cytology , Epithelium/pathology , Humans , Immunoblotting , Immunohistochemistry , Lymphatic Metastasis , Muscle, Smooth/metabolism
12.
Neoplasia ; 6(6): 777-85, 2004.
Article in English | MEDLINE | ID: mdl-15720804

ABSTRACT

Matrix metalloproteinase (MMP)-mediated degradation of the extracellular matrix is a major factor for tumor development and expansion. This study analysed MMP-10 protein expression and activity in human lung tumors of various grade, stage, and type to address the relationship between MMP-10 and tumor characteristics and to evaluate MMP-10 as a therapeutic target in non small cell lung carcinoma (NSCLC). Unlike the majority of MMPs, MMP-10 was located in the tumor mass as opposed to tumor stroma. MMP-10 protein was observed at low levels in normal human lung tissues and at significantly higher levels in all types of NSCLC. No correlation was observed between MMP-10 protein expression and tumor type, stage, or lymph node invasion. To discriminate between active and inactive forms of MMP-10 in samples of human NSCLC, we have developed an ex vivo fluorescent assay. Measurable MMP-10 activity was detected in 42 of 50 specimens of lung cancer and only 2 of 10 specimens of histologically normal lung tissue. No relationship was observed between MMP-10 activity levels and clinicopathologic characteristics. Our results suggest that MMP-10 is expressed and active at high levels in human NSCLC compared to normal lung tissues, and, as such, is a potential target for the development of novel therapeutics for lung cancer treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Metalloendopeptidases/metabolism , Animals , Biomarkers, Tumor/analysis , Cell Line, Tumor , Enzyme Activation/physiology , Humans , Immunohistochemistry , Matrix Metalloproteinase 10 , Mice , Transplantation, Heterologous
13.
Int J Oncol ; 24(1): 115-25, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14654948

ABSTRACT

A panel of tumour models used extensively for in vivo evaluation of new drugs was characterised for their p53 status. Basal p53 protein levels were measured by immunodetection on both formalin-fixed tumour tissue and from protein extracts of fresh tumours. High levels of nuclear-specific staining, indicative of p53 mutation, was seen in 15/25 tumours, with the remainder showing intermittent or no staining. The functional status of p53 cDNA from these tumours was assayed within the functional analysis of separated alleles in yeast (F.A.S.A.Y.) reporter system. The cDNA from those tumours with high levels of p53 protein showed 14/15 failing to activate the reporter gene. The cDNA from tumours with low or non-detectable p53 levels showed 8/10 with wild-type p53. Tumours were grown subcutaneously in mice (n=10). Each mouse was given maximum tolerated doses for either doxorubicin, 5-fluorouracil or cisplatin. Tumour volumes were measured daily, alongside untreated controls. The specific growth delay values for each tumour were separated into two groups, those with functional p53 (wild-type) and those without (mutant and null status). The Mann-Whitney U test was performed on the groups of data, to evaluate differences in their response on the basis of p53 status. Cisplatin was moderately active against tumours with wild-type and mutant p53 genes with no significant difference seen between both groups. However, a significant difference in specific growth delay was seen between the two groups when treated with doxorubicin or 5-fluorouracil (P=0.05), indicating a role for p53 protein in modulating the in vivo efficacy of these agents.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Tumor Suppressor Protein p53/analysis , Animals , Blotting, Western , Cisplatin/therapeutic use , Doxorubicin/therapeutic use , Female , Fluorouracil/therapeutic use , Humans , Immunohistochemistry , Mice , Mice, Inbred Strains , Mice, Nude , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Transformation, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Xenograft Model Antitumor Assays
14.
Mol Biotechnol ; 24(3): 295-301, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12777695

ABSTRACT

It is now more than a dozen years since the enzyme telomerase was discovered, and since that time, key studies have characterized the structural components of the enzyme and the associated telomeric proteins. Since the original discovery of telomerase, a clear association with cancer has been demonstrated. In normal somatic cells the telomeres at the ends of chromosomes shorten with every cell division, whereas in cancer cells telomere length is often maintained by reactivation of the enzyme telomerase. These discoveries have led to the proposal that telomerase expression can be used as a helpful marker for diagnostic and prognostic purposes in humans. Another area of research that has developed as a result of improving knowledge and understanding of the role of telomerase in malignancy is that of cancer therapeutics. This article is an introduction to the field of telomere and telomerase research, with an introduction to recent attempts to develop novel cancer treatments based on telomerase structure and function.


Subject(s)
Telomerase/metabolism , Telomere/genetics , Animals , Cell Division , Cellular Senescence/genetics , DNA Replication , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Telomerase/antagonists & inhibitors , Telomerase/chemistry , Telomerase/genetics , Telomere/ultrastructure
15.
Cancer Chemother Pharmacol ; 72(2): 417-27, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23797686

ABSTRACT

PURPOSE: Intravenously (i.v.) administered nanomedicines have the potential for tumour targeting due to the enhanced permeability and retention (EPR) effect, but in vivo tumour models are rarely calibrated with respect to functional vascular permeability and/or mechanisms controlling intratumoural drug release. Here the effect of tumour type and tumour size on EPR-mediated tumour localisation and cathepsin B-mediated drug release was studied. METHODS: Evans Blue (10 mg/kg) and an N-(2-hydroxypropyl) methacrylamide (HPMA) copolymerĀ­doxorubicin (Dox) conjugate (FCE28068) (5 mg/kg Dox-equiv) were used as probes and tumour levels (and Dox release) measured at 1 h after i.v. administration in a panel of murine and human xenograft tumours. RESULTS: Evans Blue and FCE28068 displayed similar tumour levels in the range of 2Ā­18 % dose/g at 1 h for B16F10 and L1210. Approximately half of the tumour models evaluated exhibited tumour size-dependent accumulation of FCE28068; smaller tumours had the highest accumulation. Administration of free Dox (5 mg/kg) produced tumour levels of \2.5 % dose/g independent of tumour size. Whereas the degree of EPR-mediated targeting showed *12-fold difference across the tumour models evaluated, Dox release from FCE28068 at 1 h displayed *200-fold variation. CONCLUSIONS: Marked heterogeneity was seen in terms of EPR effect and Dox release rate, underlining the need to carefully calibrate tumour models used to benchmark nanomedicines against known relevant standard agents and for optimal development of strategies for late pre-clinical and clinical development.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Cathepsin B/pharmacology , Disease Models, Animal , Nanotechnology , Acrylamides/administration & dosage , Acrylamides/pharmacokinetics , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Coloring Agents , Delayed-Action Preparations , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Evans Blue , Humans , Leukemia L1210/drug therapy , Mice , Permeability , Polyglutamic Acid , Polymers , Xenograft Model Antitumor Assays
16.
J Pharmacol Toxicol Methods ; 64(3): 226-32, 2011.
Article in English | MEDLINE | ID: mdl-21569858

ABSTRACT

INTRODUCTION: The preclinical development and clinical progression of potential anticancer agents are highly time and resource-intensive. Traditionally, promising compounds in vitro undergo further screening in xenograft models, a long process that uses large numbers of animals. In order to hasten compound progression, the hollow fiber assay (HFA) was developed by the US National Cancer Institute as an additional filtering step in drug development, bridging the gap between in vitro and xenograft compound screening. The HFA demonstrates a good correlation in terms of clinical predictivity, and has significant reduction and refinement benefits for animal usage. In addition, the assay enables the study of basic pharmacological properties of compounds under investigation. The HFA has been mainly used as a rapid in vivo cytotoxicity screen, but has also been shown to be amenable to study drug/target interactions in vivo. One of the challenges of the HFA is the small sample sizes obtained, which can limit sensitivity. METHODS: Here we specifically focus on the detection of DNA double-strand breaks, monitoring the effects of standard and novel anti-cancer agents on human lung, colon and breast cancer cell lines using immunoblotting and flow cytometry techniques for ƎĀ³-H2A.X. This presented a further challenge due to the low abundance of the target event. RESULTS: We found a good correlation between techniques in terms of rate of detection and sensitivity confirming the ability to use the HFA for detection of these specific drug-target interactions. DISCUSSION: The results demonstrate good sensitivity and predictability for drug behavior in an assay where cell number is limited. In contrast to conventional xenograft studies, this short-term assay also enables analysis of pharmacodynamic endpoints in tumor cells in vivo. Importantly, there is a significant impact on reduction and refinement of the use of animals in incorporating this assay into the drug development process.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Breaks, Double-Stranded , Drug Discovery/instrumentation , Drug Evaluation, Preclinical/instrumentation , Drug Screening Assays, Antitumor/instrumentation , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , DNA/drug effects , Doxorubicin/pharmacology , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Drug Screening Assays, Antitumor/methods , Female , Humans , Mice , Mice, Inbred BALB C
17.
Mol Med Rep ; 3(2): 309-13, 2010.
Article in English | MEDLINE | ID: mdl-21472238

ABSTRACT

Dolastatin 10, a marine natural product peptide, is now known to act as a vascular disrupting agent (VDA). These VDA properties were not known when other aspects of its promising pre-clinical profile led to initial unsuccessful clinical trials. Auristatin PYE, a synthetic analogue of dolastatin 10, has demonstrated improved activity in preliminary in vivo studies. However, as with other VDAs, tumour eradication was incomplete due to the maintenance of functional vasculature supporting the viable tumour at the periphery of the tumour xenograft, meaning that once the VDA effect subsides, the tumour regrows. One possible strategy for removing this peripheral tumour involves combining VDA therapy with another anticancer drug with a different mechanism of action. Here, we evaluated the effect of combining auristatin PYE with cisplatin in an HCT-116 human colon adenocarcinoma xenograft model. The effects on the growth of subcutaneously implanted HCT-116 xenografts in mice following intraperitoneal administration of a single dose of 4 mgkg-1 cisplatin and intravenous administration of 1 mgkg-1 auristatin PYE were evaluated compared to the effect of each agent administered alone. The effects on the functional tumour vasculature were also assessed. Statistically significant potentiation (p<0.01) was noted with a 465% growth delay for the combination group compared to the control, and 142 and 310% growth delays for the cisplatin and auristatin PYE groups, respectively. Shut down of tumour vasculature in the combination group was similar to that observed with auristatin PYE on its own. Auristatin PYE demonstrated synergistic antitumour effects when combined with cisplatin, suggesting that a combination chemotherapy regimen would be the most effective strategy when applying this new anticancer drug.

18.
Cancer Res ; 70(17): 6902-12, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20663911

ABSTRACT

Vascular disrupting agents (VDA) offer a strategy to starve solid tumors of nutrients and oxygen concomitant with tumor shrinkage. Several VDAs have progressed into early clinical trials, but their therapeutic value seems to be compromised by systemic toxicity. In this report, we describe the design and characterization of a novel VDA, ICT2588, that is nontoxic until activated specifically in the tumor by membrane-type 1 matrix metalloproteinase (MT1-MMP). HT1080 cancer cells expressing MT1-MMP were selectively chemosensitive to ICT2588, whereas MCF7 cells that did not express MT1-MMP were nonresponsive. Preferential hydrolysis of ICT2588 to its active metabolite (ICT2552) was observed in tumor homogenates of HT1080 relative to MCF7 homogenates, mouse plasma, and liver homogenate. ICT2588 activation was inhibited by the MMP inhibitor ilomastat. In HT1080 tumor-bearing mice, ICT2588 administration resulted in the formation of the active metabolite, diminution of tumor vasculature, and hemorrhagic necrosis of the tumor. The antitumor activity of ICT2588 was superior to its active metabolite, exhibiting reduced toxicity, improved therapeutic index, enhanced pharmacodynamic effect, and greater efficacy. Coadministration of ICT2588 with doxorubicin resulted in a significant antitumor response (22.6 d growth delay), which was superior to the administration of ICT2588 or doxorubicin as a single agent, including complete tumor regressions. Our findings support the clinical development of ICT2588, which achieves selective VDA targeting based on MT-MMP activation in the tumor microenvironment.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Colchicine/analogs & derivatives , Fibrosarcoma/drug therapy , Matrix Metalloproteinases, Membrane-Associated/metabolism , Matrix Metalloproteinases/metabolism , Oligopeptides/pharmacology , Thiourea/analogs & derivatives , Angiogenesis Inhibitors/pharmacokinetics , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/enzymology , Colchicine/pharmacokinetics , Colchicine/pharmacology , Doxorubicin/pharmacology , Female , Fibrosarcoma/blood supply , Fibrosarcoma/enzymology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/pathology , Oligopeptides/pharmacokinetics , Thiourea/pharmacokinetics , Thiourea/pharmacology , Tissue Distribution , Xenograft Model Antitumor Assays
20.
J Nat Prod ; 71(3): 321-4, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18154271

ABSTRACT

Sodium pancratistatin 3,4- O-cyclic phosphate ( 2) is a novel water-soluble synthetic derivative of pancratistatin ( 1), a natural alkaloid constituent of Amaryllidaceae plants, that exhibits good cytostatic and antineoplastic activity but is highly insoluble. Unlike most other natural alkaloids it does not act by binding to tubulin, and its mechanism of action has yet to be fully elucidated. Here the efficacy of 2 in a human colon adenocarcinoma model, DLD-1, and some understanding of its mode of action are investigated. Agreeing with previous studies, low cytotoxicity in vitro was seen for 2 with IC 50's of 253 and 19.7 microM for 1 and 96 h exposures, respectively. However in vivo the compound caused statistically significant tumor growth delays ( p < 0.01) at its maximum tolerated dose, and significant vascular shutdown and tumor necrosis were observed. Like 1, the compound appeared to have an unconventional mechanism of action with no effect on microtubule structure, yet causing a G 2/M block, while it was seen to disrupt mitochondrial function. The mechanism of action of 1 and 2 appears to be similar. Thus compound 2, being considerably more soluble than 1, has good potential as an anticancer agent, and further investigation is warranted.


Subject(s)
Amaryllidaceae Alkaloids/chemical synthesis , Amaryllidaceae Alkaloids/pharmacology , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Isoquinolines/chemical synthesis , Isoquinolines/pharmacology , Models, Biological , Amaryllidaceae Alkaloids/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Benzimidazoles/analysis , Cell Cycle/drug effects , Dose-Response Relationship, Drug , Fluorescent Dyes , Humans , Inhibitory Concentration 50 , Isoquinolines/chemistry , Microtubules/drug effects , Molecular Structure , Solubility , Tumor Cells, Cultured , Water
SELECTION OF CITATIONS
SEARCH DETAIL