Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Cancer Chemother Pharmacol ; 72(2): 417-27, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23797686

RESUMEN

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) copolymer­doxorubicin (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 2­18 % 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.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Catepsina B/farmacología , Modelos Animales de Enfermedad , Nanotecnología , Acrilamidas/administración & dosificación , Acrilamidas/farmacocinética , Animales , Antibióticos Antineoplásicos/farmacocinética , Antibióticos Antineoplásicos/farmacología , Antibióticos Antineoplásicos/uso terapéutico , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Colorantes , Preparaciones de Acción Retardada , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacocinética , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Azul de Evans , Humanos , Leucemia L1210/tratamiento farmacológico , Ratones , Permeabilidad , Ácido Poliglutámico , Polímeros , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Pharmacol Toxicol Methods ; 64(3): 226-32, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21569858

RESUMEN

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 γ-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.


Asunto(s)
Antineoplásicos/farmacología , Roturas del ADN de Doble Cadena , Descubrimiento de Drogas/instrumentación , Evaluación Preclínica de Medicamentos/instrumentación , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Animales , Antineoplásicos/química , Línea Celular Tumoral , ADN/efectos de los fármacos , Doxorrubicina/farmacología , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C
3.
Cancer Res ; 70(17): 6902-12, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20663911

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Colchicina/análogos & derivados , Fibrosarcoma/tratamiento farmacológico , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Oligopéptidos/farmacología , Tiourea/análogos & derivados , Inhibidores de la Angiogénesis/farmacocinética , Animales , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/enzimología , Colchicina/farmacocinética , Colchicina/farmacología , Doxorrubicina/farmacología , Femenino , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/enzimología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/enzimología , Neovascularización Patológica/patología , Oligopéptidos/farmacocinética , Tiourea/farmacocinética , Tiourea/farmacología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Med Rep ; 3(2): 309-13, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21472238

RESUMEN

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.

5.
J Nat Prod ; 71(3): 321-4, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18154271

RESUMEN

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.


Asunto(s)
Alcaloides de Amaryllidaceae/síntesis química , Alcaloides de Amaryllidaceae/farmacología , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Isoquinolinas/síntesis química , Isoquinolinas/farmacología , Modelos Biológicos , Alcaloides de Amaryllidaceae/química , Antineoplásicos Fitogénicos/química , Bencimidazoles/análisis , Ciclo Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Colorantes Fluorescentes , Humanos , Concentración 50 Inhibidora , Isoquinolinas/química , Microtúbulos/efectos de los fármacos , Estructura Molecular , Solubilidad , Células Tumorales Cultivadas , Agua
6.
Int J Oncol ; 31(2): 353-60, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17611692

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Depsipéptidos/química , Depsipéptidos/farmacología , Animales , Ciclo Celular , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Concentración 50 Inhibidora , Ratones , Ratones Desnudos , Modelos Químicos , Necrosis , Trasplante de Neoplasias
7.
Int J Oncol ; 29(6): 1493-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17088988

RESUMEN

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.


Asunto(s)
Aminoacridinas/farmacología , Bienestar del Animal , Benzodiazepinonas/farmacología , Ensayos de Selección de Medicamentos Antitumorales/métodos , Mitomicina/farmacología , Pirroles/farmacología , Animales , Procesos de Crecimiento Celular/fisiología , Ensayo Cometa , Ensayos de Selección de Medicamentos Antitumorales/ética , Femenino , Células HL-60 , Células HT29 , Humanos , Ratones , Microscopía Fluorescente/métodos , Trasplante de Neoplasias , Trasplante Heterólogo
8.
Eur J Med Chem ; 41(3): 330-9, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16458393

RESUMEN

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.


Asunto(s)
Enzimas Reparadoras del ADN/química , Diseño de Fármacos , Guanina/análogos & derivados , Guanina/química , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Purinas/química , Cristalografía por Rayos X , ADN/química , ADN/efectos de los fármacos , Guanina/farmacología , Humanos , Modelos Biológicos , Estructura Molecular , Mutación , O(6)-Metilguanina-ADN Metiltransferasa/genética , Purinas/farmacología , Especificidad por Sustrato
9.
J Nat Prod ; 68(10): 1450-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16252907

RESUMEN

The present SAR study of combretastatin A-3 (3a) focused on replacement of the 3-hydroxyl group by a series of halogens. That approach with Z-stilbenes resulted in greatly enhanced (>10-100-fold) cancer cell growth inhibition against a panel of human cancer cell lines and the murine P388 lymphocytic leukemia cell line. Synthesis of the 3-fluoro-Z-stilbene designated fluorcombstatin (11a) and its potassium 3'-O-phosphate derivative (16c) by the route 7 --> 8a --> 11a --> 14 --> 16c illustrates the general synthetic pathway. The 3'-O-phosphoric acid ester (15) of 3-bromo-Z-stilbene 13a was also converted to representative cation salts to evaluate the potential for improved aqueous solubility, and the potassium salt (16 mg/mL in water) proved most useful. The fluoro (11a), chloro (12a), and bromo (13a) halocombstatins were nearly equivalent to combretastatin A-4 (1a) as inhibitors of tubulin polymerization and of the binding of colchicine to tubulin. The tubulin binding in cell-free systems was also retained in human umbilical vein endothelial cells. All three halocombstatins retained the powerful human cancer cell line inhibitory activity of combretastatin A-4 (1a) and proved superior to combretastatin A-3 (3a). In addition, the halocombstatins targeted Gram-positive bacteria and Cryptococcus neoformans.


Asunto(s)
Antibacterianos/síntesis química , Antineoplásicos Fitogénicos/síntesis química , Hidrocarburos Halogenados/síntesis química , Estilbenos/síntesis química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacología , Cryptococcus neoformans/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Hidrocarburos Halogenados/química , Hidrocarburos Halogenados/farmacología , Leucemia P388 , Estructura Molecular , Estereoisomerismo , Estilbenos/química , Estilbenos/farmacología , Relación Estructura-Actividad , Tubulina (Proteína)/efectos de los fármacos , Células Tumorales Cultivadas
10.
Urology ; 65(4): 815-20, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15833553

RESUMEN

OBJECTIVES: To analyze matrix metalloproteinase-10 (MMP-10) expression in transitional cell carcinoma (TCC) of the bladder, evaluate the correlations between MMP-10 protein expression and clinicopathologic parameters, and address the viability of MMP-10 as a therapeutic target for TCC. MMP-mediated degradation of the extracellular matrix is an important factor in the pathogenesis of tumorigenesis and metastasis. METHODS: Using immunohistochemistry, the expression of MMP-10 was assessed using both tissue microarrays and whole sections of archival tissue specimens representative of all grades and stages of human bladder TCC (n = 60). MMP-10 expression was also assessed in histologically normal human bladder tissue (n = 10). The immunostaining results for MMP-10 expression were examined for correlations with tumor grade and stage. RESULTS: Unlike most MMPs, MMP-10 was localized primarily in the tumor mass as opposed to the tumor stroma and was detectable in all grades and stages of TCC. Significantly greater levels of MMP-10 protein were observed in superficial (pTa, pT1; n = 38) tumors than in normal bladder tissue (P = 0.01). In contrast to the proposed role of MMPs in tumor invasion, no significant difference was observed between muscle-invasive tumors (pT2 or worse; n = 22) and histologically normal bladder tissue (P = 0.50). MMP-10 expression showed no significant correlation with tumor grade. CONCLUSIONS: The data from our study showed that, unlike most MMPs, MMP-10 was not associated with tumor aggression or invasion. Our results suggest that MMP-10 protein levels are significantly greater in the earlier stages of TCC development.


Asunto(s)
Carcinoma de Células Transicionales/metabolismo , Metaloendopeptidasas/biosíntesis , Neoplasias de la Vejiga Urinaria/metabolismo , Carcinoma de Células Transicionales/química , Humanos , Metaloproteinasa 10 de la Matriz , Metaloendopeptidasas/análisis , Neoplasias de la Vejiga Urinaria/química
11.
Clin Cancer Res ; 11(2 Pt 1): 416-20, 2005 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-15701823

RESUMEN

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.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Diferenciación Celular , Neoplasias/irrigación sanguínea , Neoplasias/terapia , Neovascularización Patológica/terapia , Animales , Sustancias de Crecimiento/fisiología , Humanos , Neovascularización Patológica/fisiopatología
12.
Clin Cancer Res ; 11(3): 971-81, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15709162

RESUMEN

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.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales/tendencias , Humanos
13.
Mol Cancer Ther ; 3(11): 1451-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15542784

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Lípidos/farmacología , Liposomas/administración & dosificación , Neoplasias/metabolismo , Fosfolipasas A/metabolismo , Profármacos/administración & dosificación , Profármacos/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Antineoplásicos/toxicidad , Biomarcadores de Tumor/antagonistas & inhibidores , Biomarcadores de Tumor/metabolismo , Catálisis/efectos de los fármacos , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Éter/administración & dosificación , Éter/química , Éter/farmacología , Éter/toxicidad , Femenino , Hemólisis/efectos de los fármacos , Humanos , Hidrólisis/efectos de los fármacos , Lípidos/administración & dosificación , Lípidos/química , Lípidos/toxicidad , Ratones , Estructura Molecular , Neoplasias/enzimología , Neoplasias/patología , Especificidad de Órganos , Fosfolipasas A/antagonistas & inhibidores , Fosfolipasas A2 , Profármacos/química , Profármacos/toxicidad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Clin Cancer Res ; 10(19): 6677-85, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15475458

RESUMEN

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.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Microtúbulos/metabolismo , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Membranas Artificiales , Ratones , Ratones Endogámicos , Microscopía Confocal , Microscopía Fluorescente , Microtúbulos/efectos de los fármacos , Trasplante de Neoplasias/métodos , Paclitaxel/farmacología , Polivinilos , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Estilbenos/farmacología , Trasplante Heterólogo , Tubulina (Proteína)/análisis , Tubulina (Proteína)/metabolismo
15.
Int J Cancer ; 112(4): 577-84, 2004 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-15382038

RESUMEN

Methionine dependence is unique to cancer cells and defined as the inability to grow in a methionine-deprived environment even if supplemented with the metabolic precursor homocysteine. Cobalamin-dependent methionine synthase (MS) catalyses the formation of methionine and tetrahydrofolate from homocysteine and methyltetrahydrofolate, thus linking the methionine and folate pathways. The apparent altered methionine metabolism in methionine-dependent cancer cells suggests a role for MS, although results to date are conflicting. We have analysed key metabolites of the MS-associated transmethylation, transsulphuration and folate pathways of the methionine-dependent MAC15A tumour model as a function of tumour progression over a 10-day period. MS activity increased 2-fold from day 1 to day 10. Cysteine, homocysteine, S-adenosylmethionine and S-adenosylhomocysteine levels in tumour cytosolic fractions decreased as a function of tumour progression. Plasma cysteine levels also decreased, whilst the distribution of folates in erythrocytes was altered, with a maximum increase in methyltetrahydrofolate observed by day 5. The increasing MS activity and decreasing cysteine levels suggest an increasing methionine requirement by the tumour, whilst the induction of enzyme activity indicates that MS is not defective in the methionine-dependent MAC15A tumour. The decrease in tumour S-adenosylmethionine and S-adenosylhomocysteine levels suggests that methionine is required for some function other than cellular methylation, e.g., incorporation into protein. Overall, the results support a theory of methionine conservation in response to tumour growth, where the methionine-dependent MAC15A tumour has a higher than normal methionine requirement.


Asunto(s)
5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/farmacología , Carcinoma/fisiopatología , Metionina/metabolismo , Animales , Carcinoma/veterinaria , División Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Metilación , Ratones
16.
J Nat Prod ; 67(9): 1445-9, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15387639

RESUMEN

Bioassay-guided fractionation was applied to the cytotoxic chloroform fraction of the red alga Polysiphonia lanosa. The major compounds of the most active fraction were identified using GLC-MS analysis as lanosol (1), methyl, ethyl, and n-propyl ethers of lanosol (1a, 1b, and 1c, respectively), and aldehyde of lanosol (2), although 1b appears to be an artifact arising during the fractionation procedure. These compounds and other known bromophenols were synthesized in addition to four novel isomers (3, 3a-c). The cytotoxic activities of all the synthetic compounds were determined against DLD-1 cells using the MTT assay. Compounds with IC(50) < 20 micromol were also tested against HCT-116 cells. Compound 3c (2,5-dibromo-3,4-dihydroxybenzyl n-propyl ether) was the most active compound against both cell lines (IC(50) = 1.72 and 0.80 micromol, respectively), and its effect on the cell cycle was studied using flow cytometry.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Hidrocarburos Bromados/aislamiento & purificación , Fenoles/aislamiento & purificación , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Inglaterra , Hidrocarburos Bromados/química , Hidrocarburos Bromados/farmacología , Concentración 50 Inhibidora , Isomerismo , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Fenoles/química , Fenoles/farmacología , Rhodophyta , Células Tumorales Cultivadas
17.
Phytother Res ; 18(7): 507-10, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15305306

RESUMEN

Bioassay guided fractionation of the roots of Cyathostemma argenteum using the brine shrimp resulted in the isolation of two uncommon flavanones, 2,5-dihydroxy-7-methoxy flavanone 1 and 2,5-dihydroxy-6,7-dimethoxy flavanone 2 while the stem bark yielded the related compounds 5-hydroxy-7-methoxy flavone 3 and 5-hydroxy-6,7-dimethoxy flavone 4. The alkaloids liriodenine 5 and discretamine 6 as well as benzyl benzoate 7 were isolated from the roots and 6 was also isolated from the stembark. In cytotoxicity tests using four human breast cancer cell lines, 1 and 2 were weakly toxic to MCF-7 cells (IC(50) = 19.6 and 19.0 microM, respectively) but showed little activity against MCF-7 cells resistant to doxorubicin or against two oestrogen receptor-deficient cell lines. Compound 5, but not 6 and 7, was moderately cytotoxic against all four cell lines. These results are discussed in the context of the traditional use of C. argenteum in the treatment of breast cancer.


Asunto(s)
Annonaceae , Antineoplásicos Fitogénicos/farmacología , Fitoterapia , Extractos Vegetales/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/uso terapéutico , Artemia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral/efectos de los fármacos , Femenino , Humanos , Concentración 50 Inhibidora , Malasia , Medicina Tradicional , Extractos Vegetales/administración & dosificación , Extractos Vegetales/uso terapéutico , Aceites de Plantas/administración & dosificación , Aceites de Plantas/farmacología , Aceites de Plantas/uso terapéutico , Raíces de Plantas
18.
Neoplasia ; 6(6): 777-85, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15720804

RESUMEN

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.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Metaloendopeptidasas/metabolismo , Animales , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Activación Enzimática/fisiología , Humanos , Inmunohistoquímica , Metaloproteinasa 10 de la Matriz , Ratones , Trasplante Heterólogo
19.
Mol Cancer Ther ; 3(12): 1565-75, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15634650

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Aductos de ADN/efectos de los fármacos , Resistencia a Antineoplásicos , Tiazoles/uso terapéutico , Animales , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Ensayo Cometa , Biología Computacional , Citocromo P-450 CYP1A1/metabolismo , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Técnicas In Vitro , Membranas Artificiales , Ratones , Ratones Desnudos , Factores de Tiempo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Int J Oncol ; 24(1): 115-25, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14654948

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteína p53 Supresora de Tumor/análisis , Animales , Western Blotting , Cisplatino/uso terapéutico , Doxorrubicina/uso terapéutico , Femenino , Fluorouracilo/uso terapéutico , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos , Ratones Desnudos , Mutación , Neoplasias/metabolismo , Neoplasias/patología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crecimiento & desarrollo , Transformación Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA