Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Br J Cancer ; 109(10): 2607-18, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24129234

RESUMEN

BACKGROUND: Aurora kinases are key regulators of cell cycle and represent new promising therapeutic targets in several human tumours. METHODS: Biological relevance of Aurora kinase-A and -B was assessed on osteosarcoma clinical samples and by silencing these genes with specific siRNA in three human osteosarcoma cell lines. In vitro efficacy of two Aurora kinases-targeting drugs (VX-680 and ZM447439) was evaluated on a panel of four drug-sensitive and six drug-resistant human osteosarcoma cell lines. RESULTS: Human osteosarcoma cell lines proved to be highly sensitive to both drugs. A decreased drug sensitivity was observed in doxorubicin-resistant cell lines, most probably related to ABCB1/MDR1 overexpression. Both drugs variably induced hyperploidy and apoptosis in the majority of cell lines. VX-680 also reduced in vitro cell motility and soft-agar cloning efficiency. Drug association experiments showed that VX-680 positively interacts with all conventional drugs used in osteosarcoma chemotherapy, overcoming the cross-resistance observed in the single-drug treatments. CONCLUSION: Aurora kinase-A and -B represent new candidate therapeutic targets for osteosarcoma. In vitro analysis of the Aurora kinases inhibitors VX-680 and ZM447439 indicated in VX-680 a new promising drug of potential clinical usefulness in association with conventional osteosarcoma chemotherapeutic agents.


Asunto(s)
Antineoplásicos/uso terapéutico , Aurora Quinasas/antagonistas & inhibidores , Neoplasias Óseas/tratamiento farmacológico , Osteosarcoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adulto , Aurora Quinasas/genética , Benzamidas/uso terapéutico , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Evaluación Preclínica de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Terapia Molecular Dirigida/métodos , Osteosarcoma/genética , Osteosarcoma/patología , Piperazinas/uso terapéutico , Quinazolinas/uso terapéutico , Células Tumorales Cultivadas , Adulto Joven
2.
Br J Cancer ; 107(8): 1302-9, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-22929887

RESUMEN

BACKGROUND: Human immune system (HIS)-engrafted mice are new tools to investigate human immune responses. Here, we used HIS mice to study human immune responses against human HER-2-positive cancer cells and their ability to control tumour growth and metastasis. METHODS: BALB/c Rag2(-/-), Il2rg(-/-) mice were engrafted with CD34(+) or CD133(+) human cord blood hematopoietic stem cells (HSC) and vaccinated with human HER-2-positive cancer cells SK-OV-3 combined to human IL-12. RESULTS: Both CD34(+) or CD133(+) human HSC gave long-term engraftment and differentiation, both in peripheral blood and in lymphoid organs, and production of human antibodies. Vaccinated mice produced specific anti-HER-2 human IgG. An s.c. SK-OV-3 challenge was significantly inhibited (but not abolished) in both vaccinated and non-vaccinated HIS mice. Tumours were heavily infiltrated with human and murine cells, mice showed NK cells and production of human interferon-γ, that could contribute to tumour growth inhibition. Vaccinated HIS mice showed significantly inhibited lung metastases when compared with non-vaccinated HIS mice and to non-HIS mice, along with higher levels of tumour-infiltrating human dendritic cells. CONCLUSION: Anti-HER-2 responses were elicited through an adjuvanted allogeneic cancer cell vaccine in HIS mice. Human immune responses elicited in HIS mice effectively inhibited lung metastases.


Asunto(s)
Antígenos CD34/inmunología , Antígenos CD/inmunología , Vacunas contra el Cáncer/inmunología , Glicoproteínas/inmunología , Neoplasias Pulmonares/inmunología , Péptidos/inmunología , Receptor ErbB-2/inmunología , Antígeno AC133 , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/inmunología , Humanos , Isotipos de Inmunoglobulinas/inmunología , Neoplasias Pulmonares/secundario , Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C
3.
J Exp Med ; 194(9): 1195-205, 2001 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-11696586

RESUMEN

Transgenic Balb/c mice expressing the transforming rat HER-2/neu oncogene develop early and multifocal mammary carcinomas. Within the first 5 months of life the tissue-specific expression of HER-2/neu causes a progression in all their 10 mammary glands from atypical hyperplasia to invasive carcinoma. It was previously observed that chronic administration of interleukin (IL)-12 increased tumor latency, but every mouse eventually succumbed to multiple carcinomas. A significant improvement in tumor prevention was sought by administering allogeneic mammary carcinoma cells expressing HER-2/neu combined with systemic IL-12. This treatment reduced tumor incidence by 90% and more than doubled mouse lifetime. For the maximum prevention p185(neu) antigen must be expressed by allogeneic cells. IL-12 treatment strongly increased the cell vaccine efficacy. The mammary glands of mice receiving the combined treatment displayed a markedly reduced epithelial cell proliferation, angiogenesis, and HER-2/neu expression, while the few hyperplastic foci were heavily infiltrated by granulocytes, macrophages, and CD8(+) lymphocytes. Specific anti-HER-2/neu antibodies were produced and a nonpolarized activation of CD4(+) and CD8(+) cells secreting IL-4 and interferon (IFN)-gamma were evident. A central role for IFN-gamma in the preventive effect was proven by the lack of efficacy of vaccination in IFN-gamma gene knockout HER-2/neu transgenic Balb/c mice. A possible requirement for IFN-gamma is related to its effect on antibody production, in particular on IgG2a and IgG2b subclasses, that were not induced in IFN-gamma knockout HER-2/neu mice. In conclusion, our data show that an allogeneic HER-2/neu-expressing cell vaccine combined with IL-12 systemic treatment can prevent the onset of genetically determined tumors.


Asunto(s)
Adyuvantes Inmunológicos , Vacunas contra el Cáncer/inmunología , Interleucina-12/inmunología , Neoplasias Mamarias Experimentales/prevención & control , Receptor ErbB-2/fisiología , Animales , Mama/patología , Linfocitos T CD4-Positivos/inmunología , Trasplante de Células , Femenino , Inmunidad Celular/inmunología , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/administración & dosificación , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Ratas , Receptor ErbB-2/genética , Trasplante Homólogo , Células Tumorales Cultivadas , Vacunación/métodos
4.
J Exp Med ; 188(3): 589-96, 1998 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-9687535

RESUMEN

The ability of interleukin (IL)-12 to prevent tumors when administered to individuals with a genetic risk of cancer was studied in two lines of transgenic mice expressing rat HER-2/neu oncogene in the mammary gland. Female BALB/c (H-2(d)) mice carrying the activated HER-2/ neu oncogene show no morphological abnormalities of the mammary gland until 3 wk of age. They then progress through atypical hyperplasia to in situ lobular carcinoma and at 33 wk of age all 10 mammary glands display invasive carcinomas. Adult FVB mice (H-2(q)) carrying the HER-2/neu protooncogene develop mammary carcinomas with a longer latency (38-49 wk) and a lower multiplicity (mean of 2.6 tumors/mice). Treatment with IL-12 (5 daily intraperitoneal injections, 1 wk on, 3 wk off; the first course with 50 ng IL-12/day, the second with 100 ng IL-12/day) begun at 2 wk of age in BALB/c mice and at 21 wk of age in FVB mice markedly delayed tumor onset and reduced tumor multiplicity. Analogous results were obtained in immunocompetent and permanently CD8(+) T lymphocyte-depleted mice. In both transgenic lines, tumor inhibition was associated with mammary infiltration of reactive cells, production of cytokines and inducible nitric oxide synthase, and reduction in microvessel number, in combination with a high degree of hemorrhagic necrosis.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma in Situ/prevención & control , Carcinoma Lobular/prevención & control , Interleucina-12/farmacología , Neoplasias Mamarias Experimentales/prevención & control , Receptor ErbB-2/fisiología , Animales , Antineoplásicos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Carcinoma in Situ/genética , Carcinoma in Situ/inmunología , Carcinoma Lobular/genética , Carcinoma Lobular/inmunología , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Femenino , Interferón gamma/inmunología , Interleucina-12/inmunología , Depleción Linfocítica , Masculino , Neoplasias Mamarias Experimentales/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa de Tipo II , Ratas , Receptor ErbB-2/genética , Factor de Necrosis Tumoral alfa/inmunología , Molécula 1 de Adhesión Celular Vascular/biosíntesis
5.
Ann Oncol ; 20(2): 213-26, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18842614

RESUMEN

The epidermal growth factor receptor (EGFr) is one of the most studied molecules as a target for cancer therapy. Over these last few years, several studies attempting to identify predictive biomarkers of treatment response, such as the receptor status or other molecules related to the downstream signalling pathway, have been conducted. However, from a clinical point of view, the information obtained from ex vivo analyses still has various limitations that may be overcome by the combination with molecular imaging technologies which may provide a noninvasive, global, in vivo evaluation of the molecular tumour background. The aim of this review is to report the preclinical results of all positron emission tomography (PET) tracers synthesized until now for in vivo detection of EGFr in cancer. Two classes of PET compounds have been developed: labelled small molecules such as tyrosine kinase inhibitors and labelled monoclonal antibodies. The in vitro and in vivo results of these PET tracers are very different depending on the chemical properties, positron emission radionuclide, or animal models. As a consequence, various critical questions are still open, and the implications of a translation in the clinical setting for EGFr imaging in cancer patients is discussed.


Asunto(s)
Receptores ErbB/análisis , Neoplasias/diagnóstico por imagen , Neoplasias/enzimología , Tomografía de Emisión de Positrones/métodos , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Radioisótopos de Yodo , Células K562 , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Cintigrafía , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Curr Cancer Drug Targets ; 5(3): 221-8, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15892621

RESUMEN

Prevention of cancer through the activation of the immune system has been explored in recent years in preclinical systems thanks to the availability of several new transgenic mouse models that closely mimic the natural history of human tumors. The most thoroughly investigated model of cancer immunoprevention is the mammary carcinoma of HER-2/neu transgenic mouse. In this system it has clearly been shown that the activation of immune defences in healthy individuals can effectively prevent the subsequent onset of highly aggressive mammary carcinomas. A complete prevention was obtained using a combination of three signals (the so called "triplex" vaccine) that included the specific antigen (p185, the product of HER-2/neu) and nonspecific signals like allogeneic histocompatibility antigens and interleukin 12. The analysis of protective immune responses in models of cancer immunoprevention revealed some unexpected features, in particular the central role of antibodies in immunoprevention, at variance with conventional immuno-therapy which is firmly based on cytotoxic T cells. In the HER-2/neu system anti-p185 antibodies, in addition to immunological functions leading to tumor cell lysis, inhibit p185 dimerization and induce its internalization, resulting in the inhibition of mitogenic signaling. Most current tumor antigens appear to be unsuitable targets for cancer immunoprevention. An ideal antigen should have a crucial pathogenetic role in tumor growth to avoid the selection of antigen loss variants. Downregulation of major histocompatibility complex (MHC) expression during tumor progression frequently limits antigen recognition by MHC-restricted T cells. Thus an ideal antigen for cancer immunoprevention should be recognized both by T cells and by antibodies. Antibody binding to cell surface oncogenic determinants, in addition to complement- and cell-mediated tumor cell lysis, can block mitogenic signaling and induce internalization, resulting in tumor growth arrest. A search for new tumor antigens should be conducted among molecules that are directly involved in neoplastic transformation and are recognizable by the immune response also in MHC loss variants. Novel tumor antigens fulfilling both conditions will be crucial for the development of cancer immunoprevention and will provide new targets also for cancer immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/uso terapéutico , Neoplasias/prevención & control , Animales , Humanos , Ratones , Ratones Transgénicos
7.
Cancer Res ; 49(13): 3631-6, 1989 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-2471586

RESUMEN

The effect of various antineoplastic drugs (1-beta-D-arabinofuranosylcytosine, 5-azacytidine, cisplatin, dactinomycin, epirubicin, vincristine, and the activated metabolite of cyclophosphamide, mafosfamide) on cell differentiation in vitro was investigated using a human alveolar rhabdomyosarcoma cell clone, RMZ-RC2. These cells are able to differentiate spontaneously from small mononuclear proliferating elements to terminal, extremely elongated multinuclear structures resembling myotubes; morphological differentiation is accompanied by the expression of myosins, in particular the embryonic isoform, which was used in this study as a specific marker of myogenic differentiation. The proportion of differentiated myosin-positive cells, which was around 10-15% in control cultures 10-15 days after seeding, was increased by some drug treatments up to 30-40%; the proportion of multinuclear elements was also increased. 1-beta-D-arabinofuranosylcytosine and 5-azacytidine were the most effective drugs, while dactinomycin had no effect; other molecules ranked in between. Since significant increments were usually observed after treatment with drug doses inhibiting cell growth, the kinetic behavior of the absolute number of myosin-positive cells or nuclei was analyzed to assess whether some effects could be due to a negative selection of proliferating, undifferentiated cells. This appeared to be the case for vincristine and epirubicin, while 1-beta-D-arabinofuranosylcytosine, 5-azacytidine, and, to a lesser degree, mafosfamide and cisplatin actually seemed to increase differentiation ability.


Asunto(s)
Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Músculos/citología , Rabdomiosarcoma/patología , Azacitidina/farmacología , División Celular/efectos de los fármacos , Fusión Celular , Cisplatino/farmacología , Ciclofosfamida/farmacología , Citarabina/farmacología , Humanos , Técnicas In Vitro , Miosinas/metabolismo , Células Tumorales Cultivadas
8.
Cancer Res ; 56(19): 4503-8, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8813148

RESUMEN

Carcinoembryonic antigen (CEA) is a highly glycosylated cell surface glycoprotein belonging to the immunoglobulin superfamily. CEA has been involved in vitro in adhesion mechanisms, but little is known about the function of this glycoprotein in vivo in normal tissue differentiation and malignancy. With regard to the relationship between CEA expression and tissue differentiation, it has been reported that transfection of the CEA gene in rat L6 myoblasts results in a complete block of myogenic differentiation. To extend investigations to the transformed myogenic counterpart and examine CEA effects on differentiation and malignancy outside the colon system, we have transfected the human CEA gene in human rhabdomyosarcoma cells. Human rhabdomyosarcoma cells transfected with the CEA gene correctly expressed membrane CEA anchored via glycosylphosphatidylinositol and secreted CEA in the medium. CEA gene transfer in human rhabdomyosarcoma cells, which display a limited differentiation ability, does not further inhibit myogenic differentiation or alter in vitro proliferation or natural killer sensitivity. CEA transfection does not affect s.c. growth in nude mice, but the ectopic expression of CEA in human rhabdomyosarcoma cells can strongly inhibit their metastatic ability to lungs and adrenals after i.v. injection. The impairment of metastatic potential correlates with a reduction in the homotypic adhesion properties of the cells. These data suggest that CEA, in some systems, can interfere with intercellular adhesion and, at least for cells not metastatic to the liver, can act as an anti-metastatic molecule.


Asunto(s)
Antígeno Carcinoembrionario/fisiología , Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia/genética , Rabdomiosarcoma/genética , Neoplasias de los Tejidos Blandos/genética , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/secundario , Animales , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Diferenciación Celular , ADN Complementario/genética , Femenino , Glicosilfosfatidilinositoles/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Músculos/patología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/metabolismo , Rabdomiosarcoma/patología , Rabdomiosarcoma/secundario , Neoplasias de los Tejidos Blandos/patología , Transfección , Células Tumorales Cultivadas
9.
Cancer Res ; 58(6): 1225-30, 1998 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-9515809

RESUMEN

Immunological gene therapy of cancer relies heavily on the activation of T cells, but tumors with defects in MHC gene expression are not recognized by MHC-restricted T cells. To investigate the potential of cytokine genes for the therapy of MHC-negative tumors, we transduced B78H1, a class I-negative murine melanoma clone, with a polycistronic vector carrying murine interleukin (IL)-12 genes. The clones studied produced 400-25,000 pg/ml IL-12; their in vitro growth properties were similar to those of parental cells. A complete inhibition of growth was observed in vivo both after s.c. and i.v. administration of all IL-12 clones. IL-12-transduced cells were also used as a therapeutic vaccine in mice bearing micrometastases by nontransduced parental cells. A significant (80-90%) reduction in the number of lung nodules was obtained. Immunohistochemical analysis and studies in immunocompromised hosts showed that T cells and natural killer cells had a significant role in the elimination of IL-12-releasing cells. In situ hybridization with cytokine probes detected a strong increase in the proportion of leukocytes positive for IFN-gamma, tumor necrosis factor alpha, IL-1beta, and IFN-inducible protein 10 at the site of rejection of IL-12-engineered tumor cells. However, it was clear that the loss of in vivo growth was also due to T-cell- and natural killer cell-independent factors, possibly related to the antiangiogenic properties of IL-12. In conclusion, tumor therapy based on IL-12 gene transduction was effective on a MHC-negative metastatic tumor, suggesting a possible application to MHC-defective human neoplasms.


Asunto(s)
Antígenos H-2 , Interleucina-12/genética , Complejo Mayor de Histocompatibilidad , Melanoma Experimental/terapia , Metástasis de la Neoplasia/prevención & control , Animales , Vacunas contra el Cáncer/inmunología , Citotoxicidad Inmunológica , Terapia Genética , Huésped Inmunocomprometido , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neovascularización Patológica , Transducción Genética
10.
Cancer Res ; 56(10): 2434-9, 1996 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-8625324

RESUMEN

In osteosarcoma, resistance to chemotherapy and metastatic spread are the most important mechanisms responsible for the failure of current multimodal therapeutic programs. We have shown previously that overexpression of the MDR1 gene product P-glycoprotein is the most important predictor of an adverse clinical course in patients with osteosarcoma. treated with chemotherapy. In this study, we analyzed the relationship between P-glycoprotein expression and local aggressiveness and systemic dissemination of multidrug-resistant (MDR) human osteosarcoma cells. Compared to parental sensitive cells, MDR cells showed a decreased tumorigenicity,and metastatic ability in athymic mice, together with a reduced migratory and invasive ability and a lower homotypic adhesion ability in vitro, suggesting that P-glycoprotein overexpression is associated with a less malignant phenotype. These experimental observations were confirmed by clinical data. In fact, the time of appearance of lung metastases in a series of osteosarcoma patients treated with chemotherapy was significantly shorter in the group of cases with no expression of P-glycoprotein in the primary lesion compared to the group with P-glycoprotein overexpression. Moreover, the incidence of P-glycoprotein overexpression was found to be higher among patients with localized disease at the clinical onset than in patients with evidence of metastasis at the time of diagnosis. These data indicate that, in osteosarcoma, the MDR phenotype is not associated with a more aggressive behavior both in vitro and in clinical settings, suggesting that the previously shown association of the MDR phenotype with a worse outcome in osteosarcoma is not related to a higher metastatic ability of cells with P-glycoprotein overexpression but is more likely due to their lack of responsiveness to cytotoxic drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/fisiología , Neoplasias Óseas/patología , Resistencia a Múltiples Medicamentos/genética , Resistencia a Antineoplásicos/genética , Proteínas de Neoplasias/fisiología , Osteosarcoma/patología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Integrinas/biosíntesis , Integrinas/genética , Neoplasias Pulmonares/secundario , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/genética , Osteosarcoma/secundario
11.
Cancer Res ; 56(17): 3898-901, 1996 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8752155

RESUMEN

Human rhabdomyosarcoma cells express membrane epidermal growth factor receptor (ECF-R), which could confer responsiveness to EGF and transforming growth factor-alpha (TGF-alpha) of autocrine or paracrine origin. To study the role played by this growth factor circuit in the proliferation and differentiation of myogenic neoplastic cells, human rhabdomyosarcoma EGF-R-expressing cells (RD/18 clone) have been transfected with a plasmid containing a fragment of the EGF-R cDNA in the antisense orientation. In vitro growth and differentiative ability were studied on six antisense-transfected clones (AS) in comparison to parental RD/18 cells and to cells transfected with the plasmid containing only the neomycin resistance gene (NEO). A reduced EGF-R membrane expression was found in AS clones by decreased immunofluorescence with an anti-EGF-R monoclonal antibody. All AS transfectants had a greatly impaired proliferative ability, even when cultured in fetal bovine serum-containing medium. Proliferation of AS clones was completely blocked in medium supplemented with 2% horse serum. The differentiation ability of AS clones was heterogeneous, ranging from clones with a percentage of myosin-positive cells higher than controls to clones with a negligible myosin expression. Therefore, the growth impairment determined by the loop interruption is not sufficient to switch on the differentiation program. The role played by EGF-R in the proliferation of human rhabdomyosarcoma cells suggests that this receptor could constitute a target for a therapeutic approach.


Asunto(s)
ADN Complementario/genética , Receptores ErbB/fisiología , Oligonucleótidos Antisentido/genética , Rabdomiosarcoma/patología , Animales , Secuencia de Bases , Bovinos , Diferenciación Celular/fisiología , División Celular/fisiología , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Humanos , Datos de Secuencia Molecular , Rabdomiosarcoma/genética , Rabdomiosarcoma/metabolismo , Transfección , Células Tumorales Cultivadas
12.
Cancer Res ; 58(18): 4127-31, 1998 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-9751624

RESUMEN

Innovative, more effective treatment modalities are needed for Ewing's sarcoma (ES), a neoplasm with a disappointingly low survival rate despite the use of aggressive multimodal therapeutic approaches. We have previously shown (K. Scotlandi et al, Cancer Res., 56: 4570-4574, 1996) the existence and the pathogenetic relevance of an autocrine loop that is mediated by the insulin-like growth factor-I receptor (IGF-IR) and is crucial for the survival and proliferation of ES cells in vitro. In this study, we report that the IGF-IR-blocking monoclonal antibody alphaIR3 may also significantly inhibit ES cell growth in vivo. In particular, in almost one-half of the animals tested, after s.c. inoculation with TC-71 ES cells, the blockage of IGF-IR by alphaIR3 induced a complete regression of tumors that developed, which suggests that IGF-IR is valuable as a specific target for novel therapeutic strategies. In addition, suramin, a drug that can interfere with growth factor binding with their receptors, inhibited the tumorigenic and the metastatic ability of TC-71 cells and, therefore, is a promising agent to be combined with conventional cytotoxic drugs for the design of more effective therapeutic regimens.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Neoplasias Óseas/prevención & control , Receptores de Somatomedina/antagonistas & inhibidores , Sarcoma de Ewing/prevención & control , Suramina/uso terapéutico , Animales , Neoplasias Óseas/patología , Femenino , Ratones , Ratones Desnudos , Sarcoma de Ewing/patología , Sarcoma de Ewing/secundario , Trasplante Heterólogo
13.
Clin Cancer Res ; 4(8): 1843-9, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9717810

RESUMEN

CD40, a membrane glycoprotein of the tumor necrosis factor receptor family, is expressed by several tumor types, including B-cell lymphomas, carcinomas, and melanoma, but little is known concerning its expression by sarcoma. We used flow cytometry to analyze the expression of CD40 in human cell lines derived from 12 osteosarcomas, 6 Ewing's sarcomas, and 5 rhabdomyosarcomas. Detectable CD40 levels ranging from low to very high were found in one-third of osteosarcomas, whereas five of six Ewing's sarcomas expressed intermediate levels of CD40; all rhabdomyosarcomas were CD40-negative. At the tissue level, two of eight primary high-grade osteosarcomas showed CD40-positive immunostaining. Osteosarcoma cells and Ewing's sarcoma cells expressing CD40 were treated with recombinant soluble CD40 ligand to analyze CD40 function. Treatment with soluble CD40 ligand increased the level of apoptotic cells and stimulated the transcription of matrix metalloproteinase 9 gene, enhancing matrix metalloproteinase 9 enzyme secretion. The results indicate that in human osteosarcoma and Ewing's sarcoma, CD40 is a functional receptor whose engagement can have opposite effects on tumor cell survival and malignancy.


Asunto(s)
Neoplasias Óseas/metabolismo , Antígenos CD40/biosíntesis , Osteosarcoma/metabolismo , Sarcoma de Ewing/metabolismo , Apoptosis/efectos de los fármacos , Biopsia , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Ligando de CD40 , Colagenasas/biosíntesis , Colagenasas/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Humanos , Inmunohistoquímica , Metaloproteinasa 9 de la Matriz , Glicoproteínas de Membrana/farmacología , Osteosarcoma/enzimología , Osteosarcoma/patología , Rabdomiosarcoma/enzimología , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma/patología , Sarcoma de Ewing/enzimología , Sarcoma de Ewing/patología , Células Tumorales Cultivadas
14.
Transplant Proc ; 37(5): 2144-7, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15964362

RESUMEN

Immunosuppressive therapies associated with organ transplantation produce an increased risk of cancer development. Malignancies are increased in transplant recipients because of the impaired immune system. Moreover, experimental data point to a tumor-promoting activity of various immunosuppressive agents. In this study, we compared the effects of 4 immunosuppressive agents with different mechanisms of action (cyclosporine, rapamycin, mycophenolic acid, and leflunomide) on the in vitro growth of various tumor cell lines and umbilical vein endothelial cells. To varying degrees rapamycin (10 ng/mL), mycophenolic acid (300 nmol/L), and leflunomide (30 micromol/L) highly inhibited the growth of human rhabdomyosarcoma, hepatocellular carcinoma, colorectal carcinoma, and endothelial cells. In contrast, cyclosporine (100 ng/mL) did not affect their growth. Our data suggest that regimens containing rapamycin, mycophenolic acid, or leflunomide, which have both immunosuppressive and antitumor activities, should be preferred in transplant recipients to minimize the risk of tumors.


Asunto(s)
Antineoplásicos , Ciclosporina/farmacología , Inmunosupresores , Carcinoma Hepatocelular , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales , Humanos , Terapia de Inmunosupresión/métodos , Inmunosupresores/farmacología , Isoxazoles/farmacología , Células Jurkat , Leflunamida , Neoplasias Hepáticas , Ácido Micofenólico/farmacología , Rabdomiosarcoma , Sirolimus/farmacología
15.
Hum Gene Ther ; 6(6): 743-52, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7548274

RESUMEN

The mouse mammary carcinoma TS/A, of BALB/c (H-2d) origin, was transfected with the murine interferon-gamma (IFN-gamma) gene (Int. J. Cancer 55: 320, 1993). We used IFN-gamma transfectants as recipients for a second round of transfections with murine allogeneic class I histocompatibility (H-2b) genes that are modulated by IFN. Transfectants with either gene alone, as well as parent TS/A cells (TS/A-pc), were used as controls. Only double transfectants expressed high levels of the allogeneic H-2b genes, while in H-2b single transfectants the expression was very low (but was induced by treatment with exogenous IFN-gamma). The tumorigenic potential of IFN-gamma or H-2b single transfectants was reduced in comparison to TS/A-pc. IFN-gamma+H-2Kb double transfectants were almost nontumorigenic, while IFN-gamma+H-2Db clones gave rise to tumors in about one-half of mice. The experimental metastatic ability of all IFN-gamma+H-2b double transfectants was very low. IFN-gamma single transfectants were known to induce a strong macrophage response in the host. The expression of allogeneic H-2 antigens added a T-lymphocyte-mediated response that accounted for the lower tumorigenicity of double transfectants. These results show that it is possible to steer the immune response evoked by tumor cells for therapeutic purposes. Moreover, the high H-2 expression obtained in IFN-gamma+H-2b double transfectants suggests that single IFN-gamma transfectants are ideal recipients for all IFN-sensitive genes. This approach can be used also for other general-purpose inducers of gene expression.


Asunto(s)
Antígenos H-2/genética , Interferón gamma/genética , Neoplasias Mamarias Experimentales/genética , Metástasis de la Neoplasia/genética , Transducción Genética , Animales , Citotoxicidad Inmunológica , Femenino , Huésped Inmunocomprometido , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia/prevención & control , Transfección , Células Tumorales Cultivadas
16.
Hum Gene Ther ; 9(2): 217-24, 1998 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-9472781

RESUMEN

The parental cells of the TSA murine mammary adenocarcinoma (TSA-pc) were transfected with both the interferon-gamma (IFN-y) gene and the cytosine deaminase (CD) suicide gene to obtain a therapeutic vaccine active against TSA-pc lung metastases. Even in the absence of treatment with the prodrug 5-fluorocytosine (5-FC), the local growth of double transfectants (CD-y clones) was inhibited by a marked recruitment of granulocytes and macrophages. In mice harboring TSA-pc micrometastases, therapeutic vaccination with either IFN-gamma or CD single transfectants reduced the number of lung nodules, whereas CD-gamma double transfectants abrogated metastasis growth in up to 80% of mice. Treatment of mice with 5-FC did not alter the curative efficacy of CD-gamma double-transfectant cells. By contrast, in mice vaccinated with CD single-transfectant cells, 5-FC treatment caused a significant loss of their curative activity. Host T cells played an active role in the cure of lung metastases, because vaccination of nude mice with CD-gamma cells was uneffective.


Asunto(s)
Adenocarcinoma/genética , Interferón gamma/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Neoplasias Mamarias Experimentales/genética , Nucleósido Desaminasas/genética , Transfección , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Citosina Desaminasa , Citotoxicidad Inmunológica , Femenino , Terapia Genética , Inmunoterapia Activa , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Linfocitos T Citotóxicos/inmunología
17.
Gene ; 274(1-2): 139-49, 2001 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-11675006

RESUMEN

Rhabdomyosarcoma is a soft tissue tumor committed to the myogenic lineage but arrested prior to terminal differentiation. To identify new genes implicated in the block in myogenic differentiation of rhabdomyosarcoma cells and those responsible for their proceeding along the myogenic pathway we used cDNA microarrays to compare gene expression profiles of two clones of the human embryonal rhabdomyosarcoma cell line RD with different myogenic potentials: RD/12, which is unable to differentiate, and RD/18, which shows elements able to terminally differentiate, as defined by the expression of myosin heavy chain (up to 50% of the population) and the formation of multinucleated myotube-like structures. We identified 80 genes differentially expressed by the two clones. Differentiating RD/18 cells overexpressed the myogenic transcription factor myogenin along with known myogenic markers; myogenin transfection into undifferentiated RD/12 cells was able to revert the phenotype giving rise to 94% of clones displaying a differentiated morphology. RD/18 cells also expressed several genes not known to be expressed in rhabdomyosarcoma or muscle cells, such as pigment-epithelium derived factor and endothelin-3. Poorly differentiated RD/12 cells, along with genes related to mesenchymal lineage or early myogenic commitment, also expressed genes not previously known to be related to the differentiation block of human rhabdomyosarcoma, such as monocyte chemotactic protein-1, connective tissue growth factor and insulin-like growth factor binding protein-5. Differential expression of these genes in a time course of differentiation suggested their potential roles as either new myogenic markers or repressors of differentiation. These results identify a cluster of new genes related to the aberrant myogenic differentiation program of human rhabdomyosarcoma cells.


Asunto(s)
Diferenciación Celular/genética , Perfilación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular , Músculos/metabolismo , Rabdomiosarcoma/genética , Quimiocina CCL2/genética , Factor de Crecimiento del Tejido Conjuntivo , Relación Dosis-Respuesta a Droga , Endotelina-3/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Sustancias de Crecimiento/genética , Humanos , Proteínas Inmediatas-Precoces/genética , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/farmacología , Músculos/citología , Miogenina/genética , Miosinas/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Rabdomiosarcoma/patología , Factores de Tiempo , Células Tumorales Cultivadas
18.
Clin Exp Metastasis ; 8(2): 215-24, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2107993

RESUMEN

The effects of recombinant tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) on B16 mouse melanoma experimental metastatic ability and major histocompatibility complex (H-2b) antigens expression were studied. B16 cells exposed in vitro to TNF-alpha had an increased H-2 expression and were more metastatic than untreated cells. The simultaneous treatment with TNF-alpha and IFN-gamma amplified the enhancement of experimental metastasis and all other effects obtained with TNF-alpha alone. The B16 clone B78H1, selectively resistant to H-2 induction and to enhancement of metastatic ability by IFN-gamma, was not affected by treatment with TNF-alpha and with TNF-alpha + IFN-gamma. These findings contribute to a better understanding of the pleiotropic effects of TNF, some of which can have opposing actions in the complex tumor-host relationships.


Asunto(s)
Interferón gamma/farmacología , Metástasis de la Neoplasia , Factor de Necrosis Tumoral alfa/farmacología , Animales , Relación Dosis-Respuesta a Droga , Antígenos H-2/análisis , Células Asesinas Naturales/efectos de los fármacos , Masculino , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL
19.
Clin Exp Metastasis ; 16(2): 123-8, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9514093

RESUMEN

A spontaneously metastatic murine mammary adenocarcinoma, TSA, has been transduced with the gene for interferon alpha1 (IFN-alpha). Transfectants were used for the immunotherapy of mice bearing lung colonies induced by the intravenous inoculation of non-transduced parental cells. A significant reduction in the number of tumor colonies was obtained when repeated subcutaneous administrations of mitomycin C-blocked transfectant cells were given, commencing 3 days after an intravenous challenge with TSA cells. Intraperitoneal vaccination induced a stronger anti-tumor response than subcutaneous vaccination, and the proportion of tumor-free mice reached 50%. The potency of IFN-alpha transfectants was similar to that of IFN-gamma transfectants previously obtained from TSA. Admixture of IFN-alpha and IFN-gamma transfectant cells in the same vaccine did not increase the curative effect over that of single vaccines. In nude mice vaccination with IFN-alpha or IFN-gamma transfectants did not lead to a reduction in the number of lung colonies, indicating that an intact T cell response was required for the therapeutic effect observed in immunocompetent mice.


Asunto(s)
Adenocarcinoma/terapia , Vacunas contra el Cáncer/uso terapéutico , Interferón-alfa/inmunología , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/terapia , Mitomicina/farmacología , Adenocarcinoma/patología , Animales , Vacunas contra el Cáncer/administración & dosificación , Femenino , Inyecciones Intraperitoneales , Inyecciones Intravenosas , Inyecciones Subcutáneas , Interferón gamma/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Linfocitos T/inmunología , Transducción Genética
20.
Eur J Cancer ; 37(13): 1719-25, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11527701

RESUMEN

The expression and biological function of Nerve Growth Factor (NGF) receptors was studied in a panel of rhabdomyosarcoma cell lines derived from embryonal and alveolar histotype. All the cell lines expressed both the high affinity receptor TrkA and the low affinity receptor p75(NTR). Treatment with exogenous NGF did not considerably alter rhabdomyosarcoma cell growth or differentiation, but significantly inhibited spontaneous apoptosis as well as apoptosis, and induced by serum starvation or apoptosis induced by treatment with cycloheximide (CHX). Rhabdomyosarcoma cell lines expressed NGF and other neurotrophins and trace amounts of NGF protein were found in the supernatants of rhabdomyosarcoma cell cultures. Blocking the putative autocrine loop with an anti-NGF antibody resulted in an increase in apoptosis compared with control cultures. These data suggest that the simultaneous presence of both high and low affinity NGF receptors engaged by endogenous or exogenous NGF might contribute to the escape from apoptosis exhibited by the rhabdomyosarcoma cells.


Asunto(s)
Receptor de Factor de Crecimiento Nervioso/fisiología , Rabdomiosarcoma/patología , Apoptosis/fisiología , Humanos , Proteínas de Neoplasias/metabolismo , Factores de Crecimiento Nervioso/metabolismo , ARN Mensajero/metabolismo , Receptor de Factor de Crecimiento Nervioso/metabolismo , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Rabdomiosarcoma/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA