Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 7(47): 76613-76627, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27780936

RESUMEN

The extent of tumor oxygenation is an important factor contributing to the efficacy of radiation therapy (RTx). Interestingly, several preclinical studies have shown benefit of combining RTx with drugs that inhibit tumor blood vessel growth, i.e. angiostatic therapy. Recent findings show that proper scheduling of both treatment modalities allows dose reduction of angiostatic drugs without affecting therapeutic efficacy. We found that whilst low dose sunitinib (20 mg/kg/day) did not affect the growth of xenograft HT29 colon carcinoma tumors in nude mice, the combination with either single dose RTx (1x 5Gy) or fractionated RTx (5x 2Gy/week, up to 3 weeks) substantially hampered tumor growth compared to either RTx treatment alone. To better understand the interaction between RTx and low dose angiostatic therapy, we explored the effects of RTx on tumor angiogenesis and tissue perfusion. DCE-MRI analyses revealed that fractionated RTx resulted in enhanced perfusion after two weeks of treatment. This mainly occurred in the center of the tumor and was accompanied by increased tissue viability and decreased hypoxia. These effects were accompanied by increased expression of the pro-angiogenic growth factors VEGF and PlGF. DCE-MRI and contrast enhanced ultrasonography showed that the increase in perfusion and tissue viability was counteracted by low-dose sunitinib. Overall, these data give insight in the dynamics of tumor perfusion during conventional 2 Gy fractionated RTx and provide a rationale to combine low dose angiostatic drugs with RTx both in the palliative as well as in the curative setting.


Asunto(s)
Inhibidores de la Angiogénesis/administración & dosificación , Antineoplásicos/administración & dosificación , Neoplasias/patología , Neovascularización Patológica , Radioterapia , Animales , Línea Celular Tumoral , Quimioradioterapia , Terapia Combinada , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de la radiación , Humanos , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Hipoxia/radioterapia , Imagen por Resonancia Magnética/métodos , Ratones , Neoplasias/diagnóstico por imagen , Neoplasias/metabolismo , Neoplasias/terapia , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/radioterapia , Radioterapia/métodos , Ultrasonografía/métodos
2.
Blood ; 121(16): 3289-97, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23407548

RESUMEN

Pulmonary metastasis is a frequent cause of poor outcome in cancer patients. The formation of pulmonary metastasis is greatly facilitated by recruitment of myeloid cells, which are crucial for tumor cell survival and extravasation. During inflammation, homing of myeloid cells is mediated by endothelial activation, raising the question of a potential role for endothelial activation in myeloid cell recruitment during pulmonary metastasis. Here, we show that metastatic tumor cell attachment causes the induction of the endothelial activation markers vascular cell adhesion molecule-1 (VCAM-1) and vascular adhesion protein-1 (VAP-1). Induction of VCAM-1 is dependent on tumor cell-clot formation, decreasing upon induction of tissue factor pathway inhibitor or treatment with hirudin. Furthermore, inhibition of endothelial activation with a VCAM-1 blocking antibody or a VAP-1 small molecule inhibitor leads to reduced myeloid cell recruitment and diminished tumor cell survival and metastasis without affecting tumor cell adhesion. Simultaneous inhibition of VCAM-1 and VAP-1 does not result in further reduction in myeloid cell recruitment and tumor cell survival, suggesting that both act through closely related mechanisms. These results establish VCAM-1 and VAP-1 as mediators of myeloid cell recruitment in metastasis and identify VAP-1 as a potential target for therapeutic intervention to combat early metastasis.


Asunto(s)
Amina Oxidasa (conteniendo Cobre)/inmunología , Moléculas de Adhesión Celular/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Pulmón/patología , Células Mieloides/patología , Molécula 1 de Adhesión Celular Vascular/inmunología , Animales , Coagulación Sanguínea , Adhesión Celular , Línea Celular Tumoral , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Pulmón/inmunología , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/inmunología , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Células Mieloides/inmunología
3.
Blood ; 119(13): 3164-75, 2012 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-22327225

RESUMEN

Tissue factor (TF) expression by tumor cells correlates with metastasis clinically and supports metastasis in experimental settings. However, the precise pathways coupling TF to malignancy remain incompletely defined. Here, we show that clot formation by TF indirectly enhances tumor cell survival after arrest in the lung, during experimental lung metastasis, by recruiting macrophages characterized by CD11b, CD68, F4/80, and CX(3)CR1 (but not CD11c) expression. Genetic or pharmacologic inhibition of coagulation, by either induction of TF pathway inhibitor ex-pression or by treatment with hirudin, respectively, abrogated macrophage recruitment and tumor cell survival. Furthermore, impairment of macrophage function, in either Mac1-deficient mice or in CD11b-diphtheria toxin receptor mice in which CD11b-positive cells were ablated, decreased tumor cell survival without altering clot formation, demonstrating that the recruitment of functional macrophages was essential for tumor cell survival. This effect was independent of NK cells. Moreover, a similar population of macrophages was also recruited to the lung during the formation of a premetastatic niche. Anticoagulation inhibited their accumulation and prevented the enhanced metastasis associated with the formation of the niche. Our study, for the first time, links TF induced coagulation to macrophage recruitment in the metastatic process.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Neoplasias/patología , Nicho de Células Madre/fisiología , Tromboplastina/farmacología , Animales , Coagulación Sanguínea/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Humanos , Macrófagos/metabolismo , Macrófagos/fisiología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Monocitos/metabolismo , Monocitos/fisiología , Metástasis de la Neoplasia , Neoplasias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Nicho de Células Madre/efectos de los fármacos , Tromboplastina/metabolismo
4.
PLoS One ; 6(11): e25911, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22102855

RESUMEN

BACKGROUND: Preclinical imaging requires anaesthesia to reduce motion-related artefacts. For direct translational relevance, anaesthesia must not significantly alter experimental outcome. This study reports on the effects of both anaesthetic and carrier gas upon the uptake of [64Cu]-CuATSM, [(99m)Tc]-HL91 and [¹8F]-FMISO in a preclinical model of tumor hypoxia. METHODOLOGY/PRINCIPAL FINDINGS: The effect of carrier gas and anaesthetic was studied in 6 groups of CaNT-bearing CBA mice using [64Cu]-CuATSM, [(99m)Tc]-HL91 or [¹8F]-FMISO. Mice were anaesthetised with isoflurane in air, isoflurane in pure oxygen, with ketamine/xylazine or hypnorm/hypnovel whilst breathing air, or in the awake state whilst breathing air or pure oxygen. PET or SPECT imaging was performed after which the mice were killed for organ/tumor tracer quantitation. Tumor hypoxia was confirmed. Arterial blood gas analysis was performed for the different anaesthetic regimes. The results demonstrate marked influences on tumor uptake of both carrier gas and anaesthetic, and show differences between [(99m)Tc]-HL91, [¹8F]-FMISO and [64Cu]-CuATSM. [(99m)Tc]-HL91 tumor uptake was only altered significantly by administration of 100% oxygen. The latter was not the case for [¹8F]-FMISO and [64Cu]-CuATSM. Tumor-to-muscle ratio (TMR) for both compounds was reduced significantly when either oxygen or anaesthetics (isoflurane in air, ketamine/xylazine or hypnorm/hypnovel) were introduced. For [¹8F]-FMISO no further decrease was measured when both isoflurane and oxygen were administered, [64Cu]-CuATSM did show an additional significant decrease in TMR. When using the same anaesthetic regimes, the extent of TMR reduction was less pronounced for [64Cu]-CuATSM than for [¹8F]-FMISO (40-60% versus 70% reduction as compared to awake animals breathing air). CONCLUSIONS/SIGNIFICANCE: The use of anaesthesia can have profound effects on the experimental outcome. More importantly, all tested anaesthetics reduced tumor-hypoxia uptake. Anaesthesia cannot be avoided in preclinical studies but great care has to be taken in preclinical models of hypoxia as anaesthesia effects cannot be generalised across applications, nor disease states.


Asunto(s)
Adenocarcinoma/diagnóstico , Anestesia , Gases/análisis , Hipoxia/diagnóstico , Neoplasias Experimentales/diagnóstico , Tomografía de Emisión de Positrones , Radiofármacos/farmacocinética , Tomografía Computarizada de Emisión de Fotón Único , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Animales , Complejos de Coordinación , Radioisótopos de Cobre/farmacocinética , Femenino , Radioisótopos de Flúor/farmacocinética , Hipoxia/tratamiento farmacológico , Hipoxia/metabolismo , Ratones , Ratones Endogámicos CBA , Misonidazol/análogos & derivados , Misonidazol/farmacocinética , Músculos/efectos de los fármacos , Músculos/metabolismo , Músculos/patología , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Compuestos Organometálicos/farmacocinética , Compuestos de Organotecnecio/farmacocinética , Oxígeno/metabolismo , Fármacos Sensibilizantes a Radiaciones/farmacocinética , Tiosemicarbazonas/farmacocinética , Distribución Tisular
5.
Int J Cancer ; 129(8): 1979-89, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21154772

RESUMEN

Vascular-targeted therapeutics are increasingly used in the clinic. However, less is known about the direct response of tumor cells to these agents. We have developed a combretastatin-A-4-phosphate (CA4P) resistant variant of SW1222 human colorectal carcinoma cells to examine the relative importance of vascular versus tumor cell targeting in the ultimate treatment response. SW1222(Res) cells were generated through exposure of wild-type cells (SW1222(WT) ) to increasing CA4P concentrations in vitro. Increased resistance was confirmed through analyses of cell viability, apoptosis and multidrug-resistance (MDR) protein expression. In vivo, comparative studies examined tumor cell necrosis, apoptosis, vessel morphology and functional vascular end-points following treatment with CA4P (single 100 mg/kg dose). Tumor response to repeated CA4P dosing (50 mg/kg/day, 5 days/week for 2 weeks) was examined through growth measurement, and ultimate tumor cell survival was studied by ex vivo clonogenic assay. In vitro, SW1222(Res) cells showed reduced CA4P sensitivity, enhanced MDR protein expression and a reduced apoptotic index. In vivo, CA4P induced significantly lower apoptotic cell death in SW1222(Res) versus SW1222(WT) tumors indicating maintenance of resistance characteristics. However, CA4P-induced tumor necrosis was equivalent in both lines. Similarly, rapid CA4P-mediated vessel disruption and blood flow shut-down were observed in both lines. Cell surviving fraction was comparable in the two tumor types following single dose CA4P and SW1222(Res) tumors were at least as sensitive as SW1222(WT) tumors to repeated dosing. Despite tumor cell resistance to CA4P, SW1222(Res) response in vivo was not impaired, strongly supporting the view that vascular damage dominates the therapeutic response to this agent.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias Colorrectales/irrigación sanguínea , Neoplasias Colorrectales/tratamiento farmacológico , Estilbenos/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/análisis , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Ratones , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Flujo Sanguíneo Regional/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Clin Cancer Res ; 15(11): 3781-90, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19470729

RESUMEN

PURPOSE: The therapeutic potential of combining the prototype tumor vascular-disrupting agent combretastatin A-4 3-O-phosphate (CA-4-P) with systemic nitric oxide synthase (NOS) inhibition was investigated preclinically. EXPERIMENTAL DESIGN: Vascular response (uptake of (125)I-labeled iodoantipyrine; laser Doppler flowmetry) and tumor response (histologic necrosis; cytotoxicity and growth delay) were determined. RESULTS: Inducible NOS selective inhibitors had no effect on blood flow in the P22 rat sarcoma. In contrast, the non-isoform-specific NOS inhibitor N(omega)-nitro- l-arginine (l-NNA; 1 and 10 mg/kg i.v. or chronic 0.1 or 0.3 mg/mL in drinking water) decreased the P22 blood flow rate selectively down to 36% of control at 1 hour but did not induce tumor necrosis at 24 hours. CA-4-P, at clinically relevant doses, decreased the P22 blood flow rate down to 6% of control at 1 hour for 3 mg/kg but with no necrosis induction. However, l-NNA administration enhanced both CA-4-P-induced tumor vascular resistance at 1 hour (chronic l-NNA administration) and necrosis at 24 hours, with 45% or 80% necrosis for 3 and 10 mg/kg CA-4-P, respectively. Bolus l-NNA given 3 hours after CA-4-P was the most effective cytotoxic schedule in the CaNT mouse mammary carcinoma, implicating a particular enhancement by l-NNA of the downstream consequences of CA-4-P treatment. Repeated dosing of l-NNA with CA-4-P produced enhanced growth delay over either treatment alone in P22, CaNT, and spontaneous T138 mouse mammary tumors, which represented a true therapeutic enhancement. CONCLUSIONS: The combination of NOS inhibition with CA-4-P is a promising approach for targeting tumor vasculature, with relevance for similar vascular-disrupting agents in development.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Sarcoma Experimental/tratamiento farmacológico , Estilbenos/farmacología , Animales , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Velocidad del Flujo Sanguíneo/efectos de los fármacos , Vasos Sanguíneos/patología , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Masculino , Óxido Nítrico Sintasa/metabolismo , Nitroarginina/administración & dosificación , Nitroarginina/farmacología , Ratas , Ratas Endogámicas , Sarcoma Experimental/irrigación sanguínea , Sarcoma Experimental/patología , Estilbenos/administración & dosificación , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
7.
Cancer Res ; 68(7): 2301-11, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18381437

RESUMEN

Tubulin-binding vascular-disrupting agents (VDA) are currently in clinical trials for cancer therapy but the factors that influence tumor susceptibility to these agents are poorly understood. We evaluated the consequences of modifying tumor vascular morphology and function on vascular and therapeutic response to combretastatin-A4 3-O-phosphate (CA-4-P), which was chosen as a model VDA. Mouse fibrosarcoma cell lines that are capable of expressing all vascular endothelial growth factor (VEGF) isoforms (control) or only single isoforms of VEGF (VEGF120, VEGF164, or VEGF188) were developed under endogenous VEGF promoter control. Once tumors were established, VEGF isoform expression did not affect growth or blood flow rate. However, VEGF188 was uniquely associated with tumor vascular maturity, resistance to hemorrhage, and resistance to CA-4-P. Pericyte staining was much greater in VEGF188 and control tumors than in VEGF120 and VEGF164 tumors. Vascular volume was highest in VEGF120 and control tumors (CD31 staining) but total vascular length was highest in VEGF188 tumors, reflecting very narrow vessels forming complex vascular networks. I.v. administered 40 kDa FITC-dextran leaked slowly from the vasculature of VEGF188 tumors compared with VEGF120 tumors. Intravital microscopy measurements of vascular length and RBC velocity showed that CA-4-P produced significantly more vascular damage in VEGF120 and VEGF164 tumors than in VEGF188 and control tumors. Importantly, this translated into a similar differential in therapeutic response, as determined by tumor growth delay. Results imply differences in signaling pathways between VEGF isoforms and suggest that VEGF isoforms might be useful in vascular-disrupting cancer therapy to predict tumor susceptibility to VDAs.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/tratamiento farmacológico , Estilbenos/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Animales , Línea Celular Tumoral , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrosarcoma/metabolismo , Inmunohistoquímica , Ratones , Ratones SCID , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Isoformas de Proteínas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/genética
8.
Int J Oncol ; 30(2): 453-60, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17203228

RESUMEN

Radioimmunotherapy using 131I-A5B7, an anti-CEA antibody, in combination with the vascular disrupting agent, combretastatin A4-phosphate (CA-4-P, 200 mg/kg), has produced tumor cures in SW1222 colorectal xenografts. CA-4-P causes acute tumor blood vessel shutdown, which can be monitored in clinical trials using dynamic contrast enhanced magnetic resonance imaging (DCE-MRI). The purpose of this study was to determine the magnitude of the anti-vascular effect of CA-4-P in the SW1222 tumor, at 200 mg/kg and at lower, more clinically relevant doses, using conventional assays; relate effects to changes in DCE-MRI parameters and determine the corresponding effects on tumor retention of 131I-A5B7. The tumor vascular effects of 30, 100 and 200 mg/kg CA-4-P were determined, at 4- and 24-h post-treatment, using DCE-MRI, uptake of Hoechst 33342 for tumor vascular volume and conventional histology for necrosis. The effect of CA-4-P on tumor and normal tissue 131I-A5B7 retention was also determined. A significant reduction in tumor DCE-MRI kinetic parameters, the initial area under the contrast agent concentration time curve (IAUGC) and the transfer constant (Ktrans), was demonstrated at 4 h after CA-4-P, for all dose levels. These effects persisted for at least 24 h for the 200 mg/kg group but not for lower doses. A similar pattern was seen for vascular volume and necrosis. Despite this dose response, all three dose levels increased tumor retention of radio labeled antibody to a similar degree. These results demonstrate that moderate tumor blood flow reduction following antibody administration is sufficient to improve tumor antibody retention. This is encouraging for the combination of CA-4-P and 131I-A5B7 in clinical trials.


Asunto(s)
Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Radioinmunoterapia/instrumentación , Estilbenos/farmacología , Animales , Línea Celular Tumoral , Ensayos Clínicos como Asunto , Neoplasias Colorrectales/metabolismo , Terapia Combinada , Sinergismo Farmacológico , Humanos , Radioisótopos de Yodo/uso terapéutico , Cinética , Imagen por Resonancia Magnética , Ratones , Necrosis , Trasplante de Neoplasias , Radioinmunoterapia/métodos , Factores de Tiempo
9.
Adv Drug Deliv Rev ; 57(1): 135-52, 2005 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-15518926

RESUMEN

The blood supply of solid tumours affects the outcome of treatment via its influence on the microenvironment of tumour cells and drug delivery. In addition, tumour blood vessels are an important target for cancer therapy. Intravital microscopy of tumours growing in 'window chambers' in animal models provides a means of directly investigating tumour angiogenesis and vascular response to treatment, in terms of both the morphology of blood vessel networks and the function of individual vessels. These techniques allow repeated measurements of the same tumour. Recently, multi-photon fluorescence microscopy techniques have been applied to these model systems to obtain 3D images of the tumour vasculature, whilst simultaneously avoiding some of the problems associated with the use of conventional fluorescence microscopy in living tissues. Here, we review the current status of this work and provide some examples of its use for studying the dynamics of tumour angiogenesis and vascular function.


Asunto(s)
Anisoles/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Microscopía de Fluorescencia por Excitación Multifotónica/métodos , Neoplasias/irrigación sanguínea , Estilbenos/administración & dosificación , Tecnología Farmacéutica , Animales , Anisoles/uso terapéutico , Humanos , Microcirculación , Neoplasias/tratamiento farmacológico , Estilbenos/uso terapéutico
10.
Int J Radiat Oncol Biol Phys ; 54(5): 1491-6, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12459376

RESUMEN

PURPOSE: This overview summarizes the preclinical development of tubulin-depolymerizing agents as vascular targeting agents, leading to the identification of combretastatin A4P (CA4P). METHODS AND MATERIALS: The murine tumor CaNT was implanted s.c. in the dorsum of CBA mice. Vascular function was determined after treatment using the perfusion marker Hoechst 33342 and fluorescence microscopy. Tumor cell response was assessed by using an excision assay and by measuring the delay in growth of treated tumors. RESULTS: At doses that approximated one-half the maximum tolerated dose (MTD) in CBA mice, none of the agents evaluated-i.e., taxol, melphalan, 5-fluorouracil, doxorubicin, cisplatin, gemcitabine, and irinotecan-induced any significant reduction in perfused vascular volume within the tumor mass. In contrast, CA4P at a dose of 100 mg/kg, which approximates one-fifth the MTD, induced a greater than 80% reduction in vascular function. Although colchicine did induce vascular shutdown, this occurred only at doses approximating the MTD. Histologic evaluation demonstrated that continued growth and repopulation of the tumor mass was the result of a surviving rim of viable tumor cells at the tumor periphery. CONCLUSION: These results confirm the ability of CA4P to selectively compromise vascular function in experimental tumors, inducing extensive tumor cell death at well-tolerated doses. However, despite these effects, no growth retardation is obtained when CA4P is administered alone in a single dose. The continued growth and repopulation of the tumor mass occurs from a narrow rim of viable cells at the periphery. If, as is believed, these remaining cells are the ones most sensitive to conventional cytotoxic and macromolecular approaches, CA4P and other vascular targeting agents offer considerable potential for enhancing the effectiveness of existing and emerging cancer therapies.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neovascularización Patológica , Estilbenos/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Bencimidazoles/metabolismo , Relación Dosis-Respuesta a Droga , Ratones , Ratones Endogámicos CBA , Microscopía Fluorescente , Necrosis , Trasplante de Neoplasias , Protectores contra Radiación/uso terapéutico , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
11.
Int J Radiat Oncol Biol Phys ; 54(5): 1524-31, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12459381

RESUMEN

PURPOSE: Tumor heterogeneity necessitates the use of combined therapies. We have shown that combining antibody-directed therapy with antivascular agents converts a subcurative to a curative treatment. The purpose of this study was to investigate, by radioluminographic and microscopic techniques, the regional effects of the two complementary therapies. METHODS AND MATERIALS: Nude mice bearing colorectal tumors were injected with 125I-labeled anti-carcinoembryonic antigen antibody, and images were obtained for antibody distribution and modeling studies using radioluminography. For therapy studies, the mice were given radioimmunotherapy alone (131I-A5B7 anti-carcinoembryonic antigen antibody), the antivascular agent combretastatin A-4 3-0-phosphate (200 mg/kg), or both. Extra mice were used to study the regional tumor effects of these therapies over time: relevant histochemical procedures were performed on tissue sections to obtain composite digital microscopic images of apoptosis, blood vessels, perfusion, hypoxia, and morphology. RESULTS: Antibody distribution, modeling, and immunohistochemistry showed how radioimmunotherapy (7.4 MBq/40 microg antibody) effectively treated the outer, well-oxygenated tumor region only. Combretastatin A-4 3-0-phosphate treated the more hypoxic center, and in doing so altered the relationship between tumor parameters. CONCLUSION: The combined complementary therapies produced cures by destroying tumor regions with different pathophysiologies. Relating these regional therapeutic effects to the relevant tumor parameters microscopically allows optimization of therapy and improved translation to clinical trials.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica , Animales , Terapia Combinada , Inmunoglobulina G/metabolismo , Ratones , Ratones Desnudos , Microscopía Fluorescente , Trasplante de Neoplasias , Neoplasias/inmunología , Neoplasias/patología , Radioinmunoterapia , Factores de Tiempo , Células Tumorales Cultivadas
12.
Int J Radiat Oncol Biol Phys ; 54(5): 1532-6, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12459382

RESUMEN

PURPOSE: This study investigates the enhancement of the vascular targeting activity of the tubulin-binding agent combretastatin A4 phosphate (CA4P) by various inhibitors of nitric oxide synthases. METHODS AND MATERIALS: The syngeneic tumors CaNT and SaS growing in CBA mice were used for this study. Reduction in perfused vascular volume was measured by injection of Hoechst 33342 24 h after drug administration. Necrosis (hematoxylin and eosin stain) was assessed also at 24 h after treatment. Combretastatin A4 phosphate was synthesized by a modification of the published procedure and the nitric oxide synthase inhibitors L-NNA, L-NMMA, L-NIO, L-NIL, S-MTC, S-EIT, AMP, AMT, and L-TC, obtained from commercial sources. RESULTS: A statistically significant augmentation of the reduction in perfused vascular volume by CA4P in the CaNT tumor was observed with L-NNA, AMP, and AMT. An increase in CA4P-induced necrosis in the same tumor achieved significance with L-NNA, L-NMMA, L-NIL, and AMT. CA4P induced little necrosis in the SaS tumor, but combination with the inhibitors L-NNA, L-NMMA, L-NIO, S-EIT, and L-TC was effective. CONCLUSIONS: Augmentation of CA4P activity by nitric oxide synthase inhibitors of different structural classes supports a nitric oxide-related mechanism for this effect. L-NNA was the most effective inhibitor studied.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Inhibidores Enzimáticos/farmacología , Neovascularización Patológica , Óxido Nítrico Sintasa/antagonistas & inhibidores , Estilbenos/uso terapéutico , Animales , Bencimidazoles/farmacología , Colorantes Fluorescentes/farmacología , Ratones , Ratones Endogámicos CBA , Modelos Químicos , Necrosis , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas
13.
Cancer Res ; 62(24): 7247-53, 2002 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-12499266

RESUMEN

Physiological differences between tumor and normal vasculature provide a target for drug discovery. In particular, the immature nature of tumor vasculature may render it intrinsically sensitive to disruption by agents affecting the endothelial cell cytoskeleton, including tubulin-binding agents. In this article, we report the synthesis of a water-soluble phosphate prodrug, ZD6126, of the tubulin-binding agent N-acetylcolchinol. In vitro studies demonstrate the comparative tubulin-binding properties of the prodrug and active drug, and show the induction of pronounced, reversible changes in endothelial cell morphology at subcytotoxic doses. Neither ZD6126 nor N-acetylcolchinol showed effects on the growth of human umbilical vein endothelial cells at concentrations below 100 micro M. In contrast, changes in endothelial cell morphology were seen at much lower, noncytotoxic concentrations (0.1 micro M) of ZD6126 and more pronounced effects were seen in proliferating versus confluent endothelial cell cultures. In vivo studies were carried out using a murine tumor model (CaNT) with single administration of a dose well below the maximum tolerated dose. These studies showed a large reduction in vascular volume, induction of extensive necrosis in tumors, and a reduced tumor cell yield in a clonal excision assay, consistent with vascular rather than cytotoxic effects. A viable rim of tumor remained after single-dose administration and minimal growth delay was observed. However, well-tolerated, multiple administration regimens led to pronounced tumor-growth delay. In the human xenograft FaDu, the growth delay given by a single dose of paclitaxel was enhanced by combination with a single dose of ZD6126, and the growth delay given by the combination was greater than the sum of the growth delays from the individual treatments. These findings show that ZD6126 is a promising antivascular agent for the treatment of solid tumors.


Asunto(s)
Inhibidores de la Angiogénesis/síntesis química , Inhibidores de la Angiogénesis/farmacología , Colchicina/análogos & derivados , Endotelio Vascular/efectos de los fármacos , Neoplasias Experimentales/irrigación sanguínea , Compuestos Organofosforados/síntesis química , Compuestos Organofosforados/farmacología , Profármacos/síntesis química , Profármacos/farmacología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Inhibidores de la Angiogénesis/farmacocinética , Animales , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Bovinos , Supervivencia Celular/efectos de los fármacos , Colchicina/farmacología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/citología , Femenino , Humanos , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos CBA , Ratones SCID , Necrosis , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/patología , Neovascularización Patológica/tratamiento farmacológico , Compuestos Organofosforados/farmacocinética , Neoplasias Faríngeas/irrigación sanguínea , Neoplasias Faríngeas/tratamiento farmacológico , Neoplasias Faríngeas/patología , Profármacos/farmacocinética , Unión Proteica , Tubulina (Proteína)/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Int J Cancer ; 102(1): 70-4, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12353236

RESUMEN

Tubulin depolymerizing drugs that selectively disrupt tumour-associated vasculature have recently been identified. The lead drug in this class, combretastatin A4 phosphate (CA4P), has just completed Phase I clinical trial. Previous studies have focussed on the effects of single drug doses and have demonstrated little or no retardation of tumour growth when CA4P is used alone, but significant benefit when it is combined with conventional treatment. We have investigated the effects of multiple daily or twice daily dosing with CA4P on the vascular function, cell survival and growth of syngeneic and spontaneous breast cancers in mice. In both transplanted and spontaneous tumours significant growth retardation is observed if CA4P is administered daily (10 doses x 50 mg/kg), whereas no significant effects are seen if the same total dose (500 mg/kg) is administered as a single bolus injection. This effect is attributed, at least in part, to anti-proliferative effects on the tumour and endothelial cells, which retard the revascularisation and repopulation of the tumour core that is initially necrosed by the drug treatment. Further investigation of dose scheduling showed that the initial anti-vascular effects of CA4P are enhanced by administering the drug in 2 equal doses separated between 2 and 6 hr. The twice daily dosing schedule (25 mg/kg twice a day) produced increased growth retardation compared to the 50 mg/kg once a day schedule in the transplanted CaNT tumour. It did not do so in the spontaneous T138 tumour model. These studies indicate that the potential anti-tumour activity of CA4P when used as a single agent in clinical trials may be enhanced when used in multiple dose schedules.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Fitogénicos/farmacología , Supervivencia Celular/efectos de los fármacos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Estilbenos/farmacología , Adenocarcinoma/patología , Animales , Carcinoma Adenoescamoso/tratamiento farmacológico , Carcinoma Adenoescamoso/patología , Femenino , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos CBA , Trasplante de Neoplasias , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Anticancer Res ; 22(3): 1453-8, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12168822

RESUMEN

BACKGROUND: Tubulin depolymerizing drugs, which selectively disrupt tumour neovasculature, have recently been identified. The lead drug in this class, combretastatin A4 phosphate (CA4P), has just completed Phase I clinical trial. We have continued to synthesize and evaluate a number of combretastatins, with the aim of identifying novel agents that possess single agent activity. In the studies presented here we provide data on our lead preclinical compound and compare its antivascular and antitumour activity to that of CA4P in the murine breast adenocarcinoma CaNT. This compound, designated Oxi 4503, is the diphosphate prodrug form of combretastatin A1. RESULTS: At a dose of 1 mg/kg Oxi 4503 induced a greater than 50% reduction in functional vascular volume, which increased to 80% or more following doses of 10, 25 and 50 mg/kg. In contrast, CA4P induced approximately 40% vascular shutdown at 50 mg/kg, but had no measurable effect at 10 mg/kg. In addition to these vascular effects, Oxi 4503 at doses of 100, 200 and 400 mg/kg induced significant retardation in the growth of established CaNT tumours. No significant growth retardation was obtained with single doses of up to 400 mg/kg CA4P. CONCLUSION: In summary, these studies have identified Oxi 4503 as a preclinical development candidate with more potent antivascular and antitumour effects than CA4P when used as a single agent.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antineoplásicos/farmacología , Difosfatos/farmacología , Profármacos/farmacología , Estilbenos/farmacología , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Animales , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , Ratones
17.
Int J Exp Pathol ; 83(1): 21-38, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12059907

RESUMEN

The tumour vasculature is an attractive target for therapy. Combretastatin A-4 (CA-4) and A-1 (CA-1) are tubulin binding agents, structurally related to colchicine, which induce vascular-mediated tumour necrosis in animal models. CA-1 and CA-4 were isolated from the African bush willow, Combretum caffrum, and several synthetic analogues are also now available, such as the Aventis Pharma compound, AVE8062. More soluble, phosphated, forms of CA-4 (CA-4-P) and CA-1 (CA-1-P) are commonly used for in vitro and in vivo studies. These are cleaved to the natural forms by endogenous phosphatases and are taken up into cells. The lead compound, CA-4-P, is currently in clinical trial as a tumour vascular targeting agent. In animal models, CA-4-P causes a prolonged and extensive shut-down of blood flow in established tumour blood vessels, with much less effect in normal tissues. This paper reviews the current understanding of the mechanism of action of the combretastatins and their therapeutic potential.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Bibencilos/uso terapéutico , Neoplasias/irrigación sanguínea , Neovascularización Patológica/tratamiento farmacológico , Animales , Endotelio Vascular/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Estilbenos/farmacología
18.
NMR Biomed ; 15(2): 99-105, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11870905

RESUMEN

The effects of two anti-vascular agents, combretastatin A4 phosphate (CA4P), and 5,6-dimethylxanthenone-4-acetic acid (DMXAA), on the perfusion of two human colon adenocarcinomas implanted in SCID mice, were assessed for up to 3 h using non-invasive magnetic resonance imaging (MRI) and spectroscopy techniques (MRS). MRI measurements of GdDTPA inflow showed that treatment with CA4P had little effect on the perfusion of HT29 tumours. Localized (31)P MRS measurements also showed that the drug had no significant effect on tumour cell energy status, as assessed from the ratio of the integrals of the signals from inorganic phosphate (P(i)) and nucleoside triphosphates. However, after treatment with DMXAA, perfusion was reduced and the P(i)/NTP ratio increased, indicating that the HT29 tumour is susceptible to the action of this drug. The LS174T tumour model was susceptible to both CA4P and DMXAA, using the criteria of changes in GdDTPA inflow and P(i)/NTP ratio.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Estilbenos/uso terapéutico , Xantenos/uso terapéutico , Xantonas , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/patología , Animales , Antineoplásicos/uso terapéutico , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Medios de Contraste , Gadolinio DTPA , Humanos , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Ratones , Ratones SCID , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...