Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Ther ; 24(9): 1634-43, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27380762

RESUMEN

Prognosis of primary refractory and relapsed pediatric B-lineage acute lymphoblastic leukemia (ALL) is very poor. Relapse rates significantly correlate with persistent minimal residual disease (MRD). In MRD, favorable effector-target ratios prevail and thus this situation might be optimally suited for immunotherapy with antibodies recruiting immunological effector cells. We here report on the generation, preclinical characterization and first clinical application in B-lineage ALL of an Fc-optimized CD19 antibody. This third-generation antibody (4G7SDIE) mediated enhanced antibody-dependent cellular cytotoxicity (ADCC) against leukemic blasts with effector cells from healthy volunteers and B-lineage ALL patients. The antibody was produced in a university-owned production unit and was applied on a compassionate use basis to 14 pediatric patients with refractory and relapsed B-lineage ALL at the stage of MRD. In 10/14 patients, MRD was reduced by ≥ 1 log or below the patient-individual detection limit, and 5/14 patients have achieved ongoing complete molecular remission with a median leukemia-free survival of 428 days. Two additional patients died in complete molecular remission due to complications not related to antibody therapy. Besides profound in vivo B-cell depletion, side effects were negligible. A clinical phase 1/2 study to further assess the therapeutic activity of 4G7SDIE is in preparation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD19 , Fragmentos Fc de Inmunoglobulinas , Neoplasia Residual/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Adolescente , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD19/inmunología , Antígenos CD19/metabolismo , Línea Celular Tumoral , Niño , Preescolar , Terapia Combinada , Monitoreo de Drogas , Resistencia a Antineoplásicos , Femenino , Expresión Génica , Humanos , Fragmentos Fc de Inmunoglobulinas/inmunología , Estimación de Kaplan-Meier , Masculino , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Retratamiento , Resultado del Tratamiento , Adulto Joven
2.
Int J Oncol ; 33(4): 759-65, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18813789

RESUMEN

Transforming growth factor-beta (TGF-beta) is abundantly expressed in malignant gliomas and is crucial for the tumor micromilieu. TGF-beta not only enhances migration and invasion of glioma cells but also inhibits an effective anti-glioma immune response. TGF-beta mediates its biologic effects through interactions with TGF-beta receptors (TbetaR)-I to -III. Binding of TGF-beta leads to the activation of an intracellular signaling cascade and subsequent phosphorylation of Sma and MAD-related proteins (SMAD). Soluble TGF-beta receptors (TbetaRs) abrogate the TGF-beta effect by competing for the binding of the ligand to its receptor. Here we used adenoviral gene transfer to express TbetaR-IIs and -IIIs in human glioma cell lines. TbetaR-IIs reduced SMAD2 phosphorylation and TGF-beta-dependent reporter activity. Furthermore, it enhanced glioma cell lysis by natural killer cells. TbetaR-IIIs alone were inactive in these assays, but enhanced the effects of TbetaR-IIs. Transduction of LN-308 cells with TbetaRs markedly delayed growth of intracerebral xenografts in nude mice in vivo. These data commend TbetaRs for possible experimental therapy of gliomas.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Terapia Genética/métodos , Glioma/genética , Glioma/terapia , Proteínas Serina-Treonina Quinasas/genética , Proteoglicanos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Animales , Línea Celular Tumoral , Supervivencia Celular , Glioma/patología , Humanos , Células Asesinas Naturales/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Unión Proteica , Receptor Tipo II de Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta/metabolismo
3.
Cancer Res ; 66(7): 3852-8, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16585213

RESUMEN

Regeneration and tolerance factor (RTF) was originally identified in placenta where it is thought to be essential for fetal allograft survival. Here we report that RTF mRNA and protein are also expressed in human glioma cells in vitro and in vivo. Suppression of RTF expression by RNA interference promotes the lysis of glioma cells by natural killer (NK) and T cells in vitro. Moreover, RTF-depleted glioma cells are less tumorigenic than control cells in nude mice in vivo. Depletion of NK cells in these animals abolished this effect. RTF is thus a novel aberrantly expressed molecule which confers immune privilege to human malignant gliomas.


Asunto(s)
Glioblastoma/inmunología , Factores Supresores Inmunológicos/inmunología , Animales , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Tolerancia Inmunológica , Ratones , Ratones Desnudos , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factores Supresores Inmunológicos/biosíntesis , Factores Supresores Inmunológicos/genética , Transfección , Trasplante Heterólogo
4.
Neuro Oncol ; 9(3): 259-70, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17522330

RESUMEN

Transforming growth factor-beta (TGF-beta) is a proinvasive and immunosuppressive cytokine that plays a major role in the malignant phenotype of gliomas. One novel strategy of disabling TGF-beta activity in gliomas is to disrupt the signaling cascade at the level of the TGF-beta receptor I (TGF-betaRI) kinase, thus abrogating TGF-beta-mediated invasiveness and immune suppression. SX-007, an orally active, small-molecule TGF-betaRI kinase inhibitor, was evaluated for its therapeutic potential in cell culture and in an in vivo glioma model. The syngeneic, orthotopic glioma model SMA-560 was used to evaluate the efficacy of SX-007. Cells were implanted into the striatum of VM/Dk mice. Dosing began three days after implantation and continued until the end of the study. Efficacy was established by assessing survival benefit. SX-007 dosed at 20 mg/kg p.o. once daily (q.d.) modulated TGF-beta signaling in the tumor and improved the median survival. Strikingly, approximately 25% of the treated animals were disease-free at the end of the study. Increasing the dose to 40 mg/kg q.d. or 20 mg/kg twice daily did not further improve efficacy. The data suggest that SX-007 can exert a therapeutic effect by reducing TGF-beta-mediated invasion and reversing immune suppression. SX-007 modulates the TGF-beta signaling pathway and is associated with improved survival in this glioma model. Survival benefit is due to reduced tumor invasion and reversal of TGF-beta-mediated immune suppression, allowing for rejection of the tumor. Together, these results suggest that treatment with a TGF-betaRI inhibitor may be useful in the treatment of glioblastoma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Glioma/tratamiento farmacológico , Vigilancia Inmunológica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glioma/inmunología , Humanos , Immunoblotting , Inmunohistoquímica , Ratones , Receptor Tipo I de Factor de Crecimiento Transformador beta , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 63(24): 8996-9006, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14695218

RESUMEN

The failure of conventional cancer therapy renders glioblastoma an attractive target for immunotherapy. Tumor cells expressing ligands of the activating immunoreceptor NKG2D stimulate tumor immunity mediated by natural killer (NK), gammadelta T, and CD8(+) T cells. We report that human glioma cells express the NKG2D ligands MICA, MICB, and members of the UL16-binding protein family constitutively. However, glioma cells resist NK cell cytolysis because of high MHC class I antigen expression. Plasmid-mediated or adenovirus-mediated overexpression of MICA in glioma cells enhances their sensitivity to NK and T-cell responses in vitro and markedly delays the growth of s.c. and intracerebral LN-229 human glioma cell xenografts in nude mice and of SMA-560 gliomas in syngeneic VMDk mice. Glioma cells forming progressive tumors after implantation of stably MICA-transfected human LN-229 cells lost MICA expression, indicating a strong selection against MICA expression in vivo. Rejection of MICA-expressing SMA-560 cells in VMDk mice resulted in protective immunity to a subsequent challenge with wild-type tumor cells. Finally, the growth of syngeneic intracerebral SMA-560 tumors is inhibited by peripheral vaccination with adenovirus-mediated, MICA-infected irradiated tumor cells, and vaccination results in immune cell activation in the NK and T-cell compartments in vivo. These data commend MICA immunogene therapy as a novel experimental treatment for human malignant gliomas.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Terapia Genética/métodos , Glioma/terapia , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Receptores Inmunológicos/inmunología , Animales , Vacunas contra el Cáncer/genética , Línea Celular Tumoral , Glioma/genética , Glioma/inmunología , Glioma/metabolismo , Antígenos de Histocompatibilidad Clase I/biosíntesis , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Subfamilia K de Receptores Similares a Lectina de Células NK , Receptores Inmunológicos/biosíntesis , Receptores Inmunológicos/genética , Receptores de Células Asesinas Naturales , Linfocitos T/inmunología , Transfección , Vacunación , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cancer Res ; 64(21): 7954-61, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15520202

RESUMEN

The cytokine transforming growth factor (TGF)-beta, by virtue of its immunosuppressive and promigratory properties, has become a major target for the experimental treatment of human malignant gliomas. Here we characterize the effects of a novel TGF-beta receptor (TGF-betaR) I kinase inhibitor, SD-208, on the growth and immunogenicity of murine SMA-560 and human LN-308 glioma cells in vitro and the growth of and immune response to intracranial SMA-560 gliomas in syngeneic VM/Dk mice in vivo. SD-208 inhibits the growth inhibition of TGF-beta-sensitive CCL64 cells mediated by recombinant TGF-beta1 or TGF-beta2 or of TGF-beta-containing glioma cell supernatant at an EC(50) of 0.1 mumol/L. SD-208 blocks autocrine and paracrine TGF-beta signaling in glioma cells as detected by the phosphorylation of Smad2 or TGF-beta reporter assays and strongly inhibits constitutive and TGF-beta-evoked migration and invasion, but not viability or proliferation. Peripheral blood lymphocytes or purified T cells, cocultured with TGF-beta-releasing LN-308 glioma cells in the presence of SD-208, exhibit enhanced lytic activity against LN-308 targets. The release of interferon gamma and tumor necrosis factor alpha by these immune effector cells is enhanced by SD-208, whereas the release of interleukin 10 is reduced. SD-208 restores the lytic activity of polyclonal natural killer cells against glioma cells in the presence of recombinant TGF-beta or of TGF-beta-containing glioma cell supernatant. The oral bioavailability of SD-208 was verified by demonstrating the inhibition of TGF-beta-induced Smad phosphorylation in spleen and brain. Systemic SD-208 treatment initiated 3 days after the implantation of SMA-560 cells into the brains of syngeneic VM/Dk mice prolongs their median survival from 18.6 to 25.1 days. Histologic analysis revealed no difference in blood vessel formation, proliferation, or apoptosis. However, animals responding to SD-208 showed an increased tumor infiltration by natural killer cells, CD8 T cells, and macrophages. These data define TGF-beta receptor I kinase inhibitors such as SD-208 as promising novel agents for the treatment of human malignant glioma and other conditions associated with pathological TGF-beta activity.


Asunto(s)
Receptores de Activinas Tipo I/antagonistas & inhibidores , Antineoplásicos/farmacología , Glioma/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Glioma/inmunología , Glioma/patología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1 , Factor de Crecimiento Transformador beta2
7.
Diabetes ; 52(2): 527-35, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12540631

RESUMEN

Recent experimental work indicates that the hyperglycemia-induced increase in mesangial matrix production, which is a hallmark in the development of diabetic nephropathy, is mediated by increased expression of GLUT1. Mesangial cells stably transfected with human GLUT1 mimic the effect of hyperglycemia on the production of the extracellular matrix proteins, particularly fibronectin, when cultured under normoglycemic conditions. Our investigation of the molecular mechanism of this effect has revealed that the enhanced fibronectin production was not mediated by the prosclerotic cytokine transforming growth factor (TGF)-beta1. We found markedly increased nuclear content in Jun proteins, leading to enhanced DNA-binding activity of activating protein 1 (AP-1). AP-1 inhibition reduced fibronectin production in a dosage-dependent manner. Moreover, inhibition of classic protein kinase C (PKC) isoforms prevented both the activation of AP-1 and the enhanced fibronectin production. In contrast to mesangial cells exposed to high glucose, no activation of the hexosamine biosynthetic, p38, or extracellular signal-related kinase 1 and 2 mitogen-activated protein kinase pathways nor any increase in TGF-beta1 synthesis could be detected, which could be explained by the absence of oxidative stress in cells transfected with the human GLUT1 gene. Our data indicate that increased glucose uptake and metabolism induce PKC-dependent AP-1 activation that is sufficient for enhanced fibronectin production, but not for increased TGF-beta1 expression.


Asunto(s)
Fibronectinas/genética , Mesangio Glomerular/fisiología , Proteínas de Transporte de Monosacáridos/genética , Factor de Crecimiento Transformador beta/farmacología , Animales , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Mesangio Glomerular/efectos de los fármacos , Glucosa/metabolismo , Transportador de Glucosa de Tipo 1 , Humanos , Lactatos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Ratas , Factor de Transcripción AP-1/metabolismo , Transfección , Factor de Crecimiento Transformador beta1
8.
Neuro Oncol ; 13(2): 155-64, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21183510

RESUMEN

Death receptor targeting has emerged as one of the promising novel approaches of cancer therapy. The activation of one such prototypic death receptor, CD95 (Fas/APO-1), has remained controversial because CD95 agonistic molecules have exhibited either too strong toxicity or too little activity. The natural CD95 ligand (CD95L) is a cytokine, which needs to trimerize to mediate a cell death signal. Mega-Fas-Ligand, now referred to as APO010, is a synthetic hexameric CD95 agonist that exhibits strong antitumor activity in various tumor models. Here, we studied the effects of APO010 in human glioma models in vitro and in vivo. Compared with a cross-linked soluble CD95L or a CD95-agonistic antibody, APO010 exhibited superior activity in glioma cell lines expressing CD95 and triggered caspase-dependent cell death. APO010 reduced glioma cell viability in synergy when combined with temozolomide. The locoregional administration of APO010 induced glioma cell death in vivo and prolonged the survival of tumor-bearing mice. A further exploration of APO010 as a novel antiglioma agent is warranted.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/patología , Proliferación Celular/efectos de los fármacos , Proteína Ligando Fas/farmacología , Glioma/patología , Proteínas Recombinantes de Fusión/farmacología , Receptor fas/agonistas , Animales , Western Blotting , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/prevención & control , Proteína Ligando Fas/inmunología , Citometría de Flujo , Glioma/metabolismo , Glioma/prevención & control , Humanos , Técnicas para Inmunoenzimas , Técnicas In Vitro , Ratones , Proteínas Recombinantes de Fusión/inmunología , Células Tumorales Cultivadas , Receptor fas/metabolismo
9.
J Neurosurg ; 113(2): 280-5, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19961309

RESUMEN

OBJECT: Given the overall poor outcome with current treatment strategies in malignant gliomas, immunotherapy has been considered a promising experimental approach to glioblastoma for more than 2 decades. A cell surface molecule, CD70, may induce potent antitumor immune responses via activation of the costimulatory receptor CD27 expressed on immune effector cells. There is evidence that a soluble form of CD70 (sCD70) may exhibit biological activity, too. A soluble costimulatory ligand is attractive because it may facilitate immune activation and may achieve a superior tissue distribution. METHODS: To test the antiglioma effect of sCD70, the authors genetically modified SMA-560 mouse glioma cells to secrete the extracellular domain of CD70. They assessed the immunogenicity of the transfected cells in cocultures with immune effector cells by the determination of immune cell proliferation and the release of interferon-gamma. Syngeneic VM/Dk mice were implanted orthotopically with control or sCD70-releasing glioma cells to determine a survival benefit mediated by sCD70. Depletion studies were performed to identify the cellular mediators of prolonged survival of sCD70-releasing glioma-bearing mice. RESULTS: The authors found that ectopic expression of sCD70 enhanced the proliferation and interferon-gamma release of syngeneic splenocytes in vitro. More importantly, sCD70 prolonged the survival of syngeneic VM/Dk mice bearing intracranial SMA-560 gliomas. The survival rate at 60 days increased from 5 to 45%. Antibody-mediated depletion of CD8-positive T cells abrogates the survival advantage conferred by sCD70. CONCLUSIONS: These data suggest that sCD70 is a potent stimulator of antiglioma immune responses that depend critically on CD8-positive T cells. Soluble CD70 could be a powerful adjuvant for future immunotherapy trials for glioblastoma.


Asunto(s)
Astrocitoma/terapia , Neoplasias Encefálicas/terapia , Ligando CD27/inmunología , Ligando CD27/metabolismo , Inmunoterapia/métodos , Animales , Astrocitoma/inmunología , Astrocitoma/patología , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , División Celular/fisiología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Interferón gamma/metabolismo , Depleción Linfocítica , Ratones , Ratones Endogámicos , Invasividad Neoplásica/patología , Solubilidad , Bazo/citología , Bazo/inmunología , Tasa de Supervivencia , Transfección
10.
Int J Cancer ; 118(7): 1728-35, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16217761

RESUMEN

CD70 (CD27 ligand) promotes the expansion of primed lymphocytes by enhancing cell survival. Surprisingly, we previously observed that CD70 aberrantly expressed on human glioma cells promoted immune cell apoptosis and inhibited alloreactive lysis. Here we report that ectopic expression of CD70 in mouse glioma cells enhances apoptosis of T, B and NK cells in coculture, but nevertheless promotes glioma cell lysis by NK cells in vitro. In nude mice, CD70 expression in SMA-560 gliomas delays the glioma growth upon subcutaneous (s.c.) or intracerebral (i.c.) inoculation, suggesting a role for CD70/CD27-dependent NK cell activity in tumor surveillance. In syngeneic immunocompetent VM/Dk mice, CD70 allows the rejection of s.c. and i.c. implanted SMA-560 tumors. The tumorigenicity of CD70-expressing glioma cells is abrogated when TGF-beta signaling is blocked. Moreover, mice surviving the s.c. CD70 glioma challenge subsequently also reject wild-type glioma cells administered i.c. Similarly, CD70-expressing GL-261 gliomas are rejected in syngeneic C57BL/6 mice, while glioma growth is restored in C57BL/6 CD27(-/-) mice, suggesting that the CD70/CD27 interaction recruits a tumor-specific T-cell repertoire and induces tumor-specific memory. Altogether, these observations indicate that the net effect of aberrant CD70 expression in gliomas is immune stimulatory rather than immune paralytic and encourage its application in tumor immunotherapy.


Asunto(s)
Antígenos CD/inmunología , Apoptosis , Neoplasias Encefálicas/inmunología , Glioma/inmunología , Células Asesinas Naturales/fisiología , Proteínas de la Membrana/inmunología , Factores de Necrosis Tumoral/inmunología , Animales , Antígenos CD/biosíntesis , Linfocitos B , Neoplasias Encefálicas/patología , Ligando CD27 , Glioma/patología , Humanos , Inmunoterapia , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Linfocitos T/fisiología , Factor de Crecimiento Transformador beta/fisiología , Factores de Necrosis Tumoral/biosíntesis
11.
Eur J Immunol ; 36(10): 2807-16, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17013976

RESUMEN

Stabilized synthetic RNA oligonucleotides (ORN) and protected messenger RNA (mRNA) were recently discovered to possess an immunostimulatory capacity through their recognition by TLR 7 and 8. We wanted to find out whether this danger signal is capable of triggering anti-tumor immunity when injected locally into an established tumor. Using the mouse glioma tumor cell line SMA-560 in syngenic VM/Dk mice, we were able to show that intra-tumor injections of protamine-stabilized mRNA do indeed induce tumor regression and long-term anti-tumor immunity. Residual RNA-injected tumors show CD8 infiltration. Distant injections of protamine-protected mRNA and intra-tumor injection of naked mRNA also result in anti-tumor immunity. Although they are strong danger signals, RNA are labile molecules with a short half-life: they do not trigger side effects such as long-term, uncontrolled immunostimulation evidenced by splenomegaly in CpG DNA-treated mice. In conclusion, RNA molecules are potent and safe danger signals that are relevant for active immunotherapy strategies aimed at the eradication of solid tumors.


Asunto(s)
Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , ARN Mensajero/inmunología , ARN Mensajero/uso terapéutico , Animales , Línea Celular Tumoral , Islas de CpG/inmunología , ADN/inmunología , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Ratones , Protaminas/inmunología
12.
J Neurooncol ; 66(1-2): 147-54, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15015780

RESUMEN

Subcutaneous vaccination using granulocyte-macrophage colony-stimulating factor (GM-CSF)-transduced glioma cells substantially prolongs survival in the mouse GL261 glioma model. To potentiate the efficacy of GM-CSF-based vaccination, syngeneic C57BL/6 mice bearing pre-implanted intracerebral GL261 gliomas were vaccinated twice subcutaneously with various combinations of glioma cells retrovirally engineered to release GM-CSF, interleukin (IL)-4 or macrophage inflammatory protein (MIP)-1alpha. More than 80% of the animals vaccinated with GM-CSF-secreting or GM-CSF- and IL-4-secreting cells were long-term survivors (> 120 days). Their survival was significantly prolonged compared with animals vaccinated with wild-type cells, which died after a median survival time of 30 days. The combination of IL-4 with GM-CSF did not provide a survival advantage over GM-CSF alone, regardless of whether the animals carried a small or large intracranial tumor load. Further, when the animals were vaccinated with a mixture of GM-CSF-, IL-4- and MIP-1alpha-secreting cells, the median survival was 37 days, and only 22% of the animals in this group were long-term survivors, similar to the vaccination effect of non-modified glioma cells. Thus, unexpectedly, the co-expression of MIP-1alpha, which was meant to attract T cells for stimulation by GM-CSF- and IL-4-stimulated dendritic cells, nullified the induction of an immune response against the GL261 glioma by a GM-CSF- and IL-4-expressing subcutaneous vaccine.


Asunto(s)
Neoplasias Encefálicas/terapia , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/antagonistas & inhibidores , Glioma/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Proteínas Inflamatorias de Macrófagos/administración & dosificación , Vacunación , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Quimiocina CCL3 , Quimiocina CCL4 , Sinergismo Farmacológico , Vectores Genéticos , Glioma/patología , Inmunohistoquímica , Terapia de Inmunosupresión , Inyecciones Subcutáneas , Interleucina-4/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Retroviridae/genética , Análisis de Supervivencia , Transducción Genética , Insuficiencia del Tratamiento
13.
Ann Neurol ; 54(3): 388-92, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12953273

RESUMEN

Macrophages are thought to represent a first line of defense in anti-tumor immunity. Despite infiltration by microglial cells, however, malignant gliomas are still highly aggressive tumors. We here identify monocyte chemoattractant protein-1 (MCP-1) as a critical chemoattractant for glioma-infiltrating microglial cells. MCP-1-transfected rat CNS-1 gliomas were massively infiltrated by microglial cells. Whereas MCP-1 did not promote the growth of CNS-1 cells in vitro, intracerebral CNS-1-transfected tumors grew more aggressively than control-transfected tumors. This provides the first functional evidence that MCP-1 recruits microglial cells to gliomas and promotes their growth in vivo. Microglial cells may support rather than suppress glioma growth.


Asunto(s)
Quimiocina CCL2/fisiología , Microglía/metabolismo , Animales , Quimiotaxis , Cuerpo Estriado/trasplante , Medios de Cultivo Condicionados/farmacología , Femenino , Glioma/metabolismo , Invasividad Neoplásica , Trasplante de Neoplasias , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , Ratas , Ratas Wistar , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA