Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 302
Filtrar
1.
Oncogene ; 33(23): 3014-23, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-23831570

RESUMEN

One of the hallmarks of malignancy is the polarization of tumor-associated macrophages (TAMs) from a pro-immune (M1-like) phenotype to an immune-suppressive (M2-like) phenotype. However, the molecular basis of the process is still unclear. MicroRNA (miRNA) comprises a group of small, non-coding RNAs that are broadly expressed by a variety of organisms and are involved in cell behaviors such as suppression or promotion of tumorigenesis. Here, we demonstrate that miR-19a-3p, broadly conserved among vertebrates, was downregulated in RAW264.7 macrophage cells of the M2 phenotype in conditoned medium of 4T1 mouse breast tumor cells. This downregulation correlated with an increased expression of the Fra-1 gene, which was reported to act as a pro-oncogene by supporting the invasion and progression of breast tumors. We found significant upregulation of miR-19a-3p in RAW264.7 macrophages after transfection with a miR-19a-3p mimic that resulted in a significant suppression of the expression of this gene. In addition, we could measure the activity of binding between miR-19a-3p and Fra-1 with a psiCHECK luciferase reporter system. Further, transfection of RAW264.7 macrophage cells with the miR-19a-3p mimic decreased the expression of the Fra-1 downstream genes VEGF, STAT3 and pSTAT3. Most importantly, the capacity of 4T1 breast tumor cells to migrate and invade was impaired in vivo by the intratumoral injection of miR-19a-3p. Taken together, these findings indicate that miR-19a-3p is capable of downregulating the M2 phenotype in M2 macrophages and that the low expression of this miRNA has an important role in the upregulation of Fra-1 expression and induction of M2 macrophage polarization.


Asunto(s)
Antineoplásicos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/patología , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Invasividad Neoplásica , Metástasis de la Neoplasia , Fenotipo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-fos/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección , Microambiente Tumoral , Células U937
2.
Br J Cancer ; 104(9): 1410-7, 2011 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-21468047

RESUMEN

BACKGROUND: The cancer stem cell hypothesis suggests that neoplastic clones are maintained exclusively by a small subpopulation of cells, which have indefinite proliferation and differentiation potentials and give rise to phenotypically diverse cancer cells. Cancer stem cells have been isolated by their ability to efflux Hoechst 33342 dye and are referred to as the 'side population' (SP). METHODS AND RESULTS: The Hoechst efflux assay was used to isolate and characterize the SP from murine D121 lung carcinoma cells. Here, we demonstrated that D121-SP cells contain cancer stem cell characteristics, that is, upregulation of the transcription factors SOX2 and Oct 4 in D121-SP cells. In addition, the migration of D121-SP was decreased, and apoptosis of D121-SP was upregulated following knocking down of SOX2 in D121 cells. Importantly, downregulation of SOX2 in D121 cells markedly suppressed their metastatic potential in syngeneic mice. CONCLUSIONS: These results suggest that the SP is an enriched source of lung tumour cells with stem cell properties and that SOX2 has an important role in maintaining stem cell properties and functions that may be a potential target for effective lung cancer therapy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Apoptosis , Bencimidazoles , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Citometría de Flujo , Colorantes Fluorescentes , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen/métodos , Silenciador del Gen , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factor 3 de Transcripción de Unión a Octámeros , ARN Interferente Pequeño , Receptores Notch , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética , Transducción de Señal , Regulación hacia Arriba , Proteínas Wnt
3.
Oncogene ; 29(5): 662-73, 2010 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-19966854

RESUMEN

A growing body of evidence indicates that interactions between neoplastic cells and tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) are crucial in promoting tumor cell invasion and progression. Macrophages have an ambiguous role in these processes as this M1 phenotype correlates with tumoricidal capacity, whereas TAMs of M2 phenotype exert tumor-promoting effects. In this study, we provide evidence that interactions between mouse breast tumor cells and TAMs remodel the TME, leading to the upregulation of Fra-1, a member of the FOS family of transcription factor. In turn, this proto-oncogene initiates activation of the IL-6/JAK/Stat3 signaling pathway. This creates a malignant switch in breast tumor cells, leading to increased release of proangiogenic factors MMP-9, vascular endothelial growth factor and transforming growth factor-beta from tumor cells and intensified invasion and progression of breast cancer. Proof of the concept for the crucial role played by transcription factor Fra-1 in regulating these processes was established by specific knockdown of Fra-1 with small interfering RNA, which resulted in a marked suppression of tumor cell invasion, angiogenesis and metastasis in a mouse breast cancer model. Such a strategy could eventually lead to future efficacious treatments of metastatic breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Macrófagos/metabolismo , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/fisiología , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Comunicación Celular/fisiología , Separación Celular , Progresión de la Enfermedad , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Ratones , Invasividad Neoplásica/inmunología , Invasividad Neoplásica/patología , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/inmunología , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Breast Dis ; 20: 81-91, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15687710

RESUMEN

Two novel oral DNA-based vaccines provide immune protection against breast cancer in mouse model systems. These vaccines are delivered by attenuated Salmonella typhimurium to secondary lymphoid organs and are directed against novel targets such as transcription factor Fos-related antigen 1 (Fra-1) and endoglin (CD105). Both vaccines elicit suppression of angiogenesis in the breast tumor vasculature and break peripheral tolerance by eliciting potent cell-mediated protective immunity against these tumor self-antigens resulting in effective suppression of breast tumor growth and metastasis.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/secundario , Vacunas contra el Cáncer/inmunología , Neovascularización Patológica/inmunología , Vacunas de ADN/inmunología , Animales , Neoplasias de la Mama/terapia , Humanos , Inmunoterapia
5.
Cancer Res ; 61(23): 8498-503, 2001 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-11731434

RESUMEN

The induction of a CTL response capable of eradicating disseminated tumor metastases and the establishment of a persistent tumor-protective immunity remain major goals of cancer immunotherapy. Here, we demonstrate for the first time that the combination of interleukin 2 (IL-2) targeted to the tumor microenvironment by a recombinant antibody-IL-2 fusion protein (huKS1/4-IL-2) with gene therapy by the murine chemokine MIG (CXCL9) markedly reduced s.c. tumor burden and decisively suppressed dissemination of experimental lung metastases of CT26-KSA colon carcinoma in syngeneic BALB/c mice. This combined therapy significantly prolonged the life span of these mice 3-4-fold by concurrently delivering MIG and IL-2 to the tumor site and thereby achieving chemoattraction of T cells together with their activation. The antitumor effect obtained was mediated predominantly by MHC class I antigen-restricted CD8(+) T cells with help from MHC class II antigen-restricted CD4(+) T lymphocytes. In addition, the MIG chemokine also induced angiostatic effects in the tumor vasculature. Taken together, this combination of MIG chemokine gene therapy with tumor-targeted cytokine IL-2 provides an approach for the rational design of novel cancer immunotherapy modalities.


Asunto(s)
Moléculas de Adhesión Celular , Quimiocinas CXC/genética , Neoplasias del Colon/terapia , Terapia Genética/métodos , Inmunotoxinas/farmacología , Péptidos y Proteínas de Señalización Intercelular , Interleucina-2/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Antígenos de Neoplasias/inmunología , Células COS , División Celular/efectos de los fármacos , Quimiocina CXCL9 , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Terapia Combinada , Molécula de Adhesión Celular Epitelial , Femenino , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C
6.
J Immunol ; 167(8): 4560-5, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11591784

RESUMEN

A carcinoembryonic Ag (CEA)-based DNA vaccine encoding both CEA and CD40 ligand trimer achieved effective tumor-protective immunity against murine colon carcinoma in CEA-transgenic mice by activating both naive T cells and dendritic cells. Peripheral T cell tolerance to CEA was broken in a prophylactic model by this novel, dual-function DNA vaccine, whose efficacy was further enhanced by boosts with a recombinant Ab-IL-2 fusion protein (huKS1/4-IL-2). These conclusions are supported by four lines of evidence. First, a lethal challenge of MC38-CEA-KS Ag murine colon carcinoma cells was for the first time completely rejected in 100% of experimental animals treated by oral gavage of this DNA vaccine carried by attenuated Salmonella typhimurium, followed by five boosts with huKS1/4-IL-2. Second, specific activation of dendritic cells was indicated by their marked up-regulation in expression of costimulatory molecules B7.1 (CD80), B7.2 (CD86), and ICAM-1. Third, a decisive increase over control values was observed in both MHC class I Ag-restricted cytotoxicity of CTLs from successfully vaccinated mice and secretion of proinflammatory cytokines IFN-gamma and IL-12. Fourth, activation of CTLs was augmented, as indicated by up-regulation of activity markers LFA-1, CD25, CD28, and CD69. Taken together, these results suggest that a dual-function DNA vaccine encoding CEA and CD40 ligand trimer combined with tumor-targeted IL-2 may be a promising strategy for the rational development of DNA-based cancer vaccines for future clinical applications.


Asunto(s)
Ligando de CD40/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/uso terapéutico , Carcinoma/prevención & control , Moléculas de Adhesión Celular , Neoplasias del Colon/prevención & control , Vacunas de ADN/uso terapéutico , Animales , Antígenos CD/biosíntesis , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Antígeno B7-1/biosíntesis , Antígeno B7-2 , Ligando de CD40/genética , Ligando de CD40/inmunología , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Molécula de Adhesión Celular Epitelial , Interleucina-2/genética , Interleucina-2/inmunología , Interleucina-2/uso terapéutico , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Transgénicos , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Citotóxicos/inmunología , Vacunación
7.
Vaccine ; 20(3-4): 421-9, 2001 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-11672905

RESUMEN

A DNA vaccine encoding human carcinoembryonic antigen (CEA) broke peripheral T-cell tolerance toward this tumor self-antigen expressed by Lewis lung carcinoma stably transduced with CEA in C57BL/6J mice transgenic for CEA. This vaccine, delivered by oral gavage with an attenuated strain of Salmonella typhimurium (SL7207), and boosted with an antibody-IL2 fusion protein, induced tumor-protective immunity mediated by MHC class I antigen-restricted CD8(+) T cells, resulting in eradication of subcutaneous tumors in 100% of mice and prevention of experimental pulmonary metastases in 75% of experimental animals. Both CTL and antigen-presenting dendritic cells were activated as indicated by a decisive increase in their respective activation markers CD2, CD25, CD28 as well as CD48 and CD80. The antitumor effects of this CEA-based DNA vaccine obtained in prophylactic settings, suggest that this approach could lead to the rational design of effective treatment modalities for human lung cancer.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Vacunas de ADN/inmunología , Administración Oral , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Carcinoma Pulmonar de Lewis/prevención & control , Carcinoma Pulmonar de Lewis/secundario , Células Dendríticas/fisiología , Antígenos de Histocompatibilidad Clase I/inmunología , Interleucina-12/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Vacunación , Vacunas de ADN/administración & dosificación
8.
Cancer Res ; 61(16): 6178-84, 2001 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-11507070

RESUMEN

We demonstrate that a mouse-human chimeric anti-ganglioside GD2-interleukin (IL)-2 fusion protein (ch14.18-IL2) substantially amplifies tumor-protective immunity against murine melanoma induced by an autologous oral DNA vaccine containing the murine ubiquitin gene fused to murine melanoma peptide epitopes gp100(25-35) and TRP-2(181-188). This combination therapy led to the complete rejection of a lethal challenge with B78D14 murine melanoma cells in six of eight mice and a marked suppression of s.c. tumor growth in the two remaining animals. The tumor-protective immunity was mediated by MHC class I antigen- restricted CD8(+) T cells together with CD4(+) T cell help, which was required only for tumor cell killing in the effector phase of the immune response. A single oral vaccination with the DNA vaccine, which was carried by attenuated Salmonella typhimurium, was equally as effective as three such vaccinations applied at 2-week intervals. The immunological mechanisms involved in this antitumor effect were suggested by a decisively increased secretion of tumor necrosis factor alpha TNFTnTNa and IFN-gamma from CD4(+) and CD8(+) T cells and a markedly up-regulated expression on CD8(+) T cells of high-affinity IL-2 receptor alpha chain (CD25), costimulatory molecule CD28, and adhesion molecule lymphocyte function-associated antigen-2 (LFA-2/CD2). Additionally, the combination therapy induced increased expression of costimulatory molecules B7.1 and CD48 on murine antigen-presenting cells. Taken together, our results suggest that IL-2 targeted to the tumor microenvironment by a specific antibody-IL-2 fusion protein is a potent enhancer of tumor-protective immunity induced by an oral DNA vaccine that may ultimately enhance the chances of success in its clinical application.


Asunto(s)
Inmunotoxinas/inmunología , Interleucina-2/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/prevención & control , Vacunas de ADN/inmunología , Administración Oral , Animales , Antígeno B7-1/biosíntesis , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Sinergismo Farmacológico , Epítopos/inmunología , Femenino , Gangliósidos/inmunología , Humanos , Inmunotoxinas/administración & dosificación , Interferón gamma/metabolismo , Interleucina-2/administración & dosificación , Oxidorreductasas Intramoleculares/inmunología , Activación de Linfocitos/inmunología , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Neoplasias/inmunología , Fragmentos de Péptidos/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Vacunas de ADN/administración & dosificación , Antígeno gp100 del Melanoma
9.
Proc Natl Acad Sci U S A ; 98(13): 7528-33, 2001 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-11404472

RESUMEN

Effective chemotherapy remains a key issue for successful cancer treatment in general and neuroblastoma in particular. Here we report a chemotherapeutic strategy based on catalytic antibody-mediated prodrug activation. To study this approach in an animal model of neuroblastoma, we have synthesized prodrugs of etoposide, a drug widely used to treat this cancer in humans. The prodrug incorporates a trigger portion designed to be released by sequential retro-aldol/retro-Michael reactions catalyzed by aldolase antibody 38C2. This unique prodrug was greater than 10(2)-fold less toxic than etoposide itself in in vitro assays against the NXS2 neuroblastoma cell line. Drug activity was restored after activation by antibody 38C2. Proof of principle for local antibody-catalyzed prodrug activation in vivo was established in a syngeneic model of murine neuroblastoma. Mice with established 100-mm3 s.c. tumors who received one intratumoral injection of antibody 38C2 followed by systemic i.p. injections with the etoposide prodrug showed a 75% reduction in s.c. tumor growth. In contrast, injection of either antibody or prodrug alone had no antitumor effect. Systemic injections of etoposide at the maximum tolerated dose were significantly less effective than the intratumoral antibody 38C2 and systemic etoposide prodrug combination. Significantly, mice treated with the prodrug at 30-fold the maximum tolerated dose of etoposide showed no signs of prodrug toxicity, indicating that the prodrug is not activated by endogenous enzymes. These results suggest that this strategy may provide a new and potentially nonimmunogenic approach for targeted cancer chemotherapy.


Asunto(s)
Anticuerpos Catalíticos/metabolismo , Etopósido/farmacocinética , Etopósido/toxicidad , Profármacos/farmacocinética , Profármacos/toxicidad , Animales , Biotransformación , Catálisis , División Celular/efectos de los fármacos , Fructosa-Bifosfato Aldolasa/inmunología , Cinética , Ratones , Estructura Molecular , Neuroblastoma , Profármacos/síntesis química , Factores de Tiempo , Células Tumorales Cultivadas
10.
J Immunol ; 166(11): 6944-51, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11359856

RESUMEN

The successful induction of T cell-mediated protective immunity against poorly immunogenic malignancies remains a major challenge for cancer immunotherapy. Here, we demonstrate that the induction of tumor-protective immunity by IL-12 in a murine neuroblastoma model depends entirely on the CXC chemokine IFN-gamma-inducible protein 10 (IP-10). This was established by in vivo depletion of IP-10 with mAbs in mice vaccinated against NXS2 neuroblastoma by gene therapy with a linearized, single-chain (sc) version of the heterodimeric cytokine IL-12 (scIL-12). The efficacy of IP-10 depletion was indicated by the effective abrogation of scIL-12-mediated antiangiogenesis and T cell chemotaxis in mice receiving s.c. injections of scIL-12-producing NXS2 cells. These findings were extended by data demonstrating that IP-10 is directly involved in the generation of a tumor-protective CD8+ T cell-mediated immune response during the early immunization phase. Four lines of evidence support this contention: First, A/J mice vaccinated with NXS2 scIL-12 and depleted of IP-10 by two different anti-IP-10 mAbs revealed an abrogation of systemic-protective immunity against disseminated metastases. Second, CD8+ T cell-mediated MHC class I Ag-restricted tumor cell lysis was inhibited in such mice. Third, intracellular IFN-gamma expressed by proliferating CD8+ T cells was substantially inhibited in IP-10-depleted, scIL-12 NXS2-vaccinated mice. Fourth, systemic tumor protective immunity was completely abrogated in mice depleted of IP-10 in the early immunization phase, but not if IP-10 was depleted only in the effector phase. These findings suggest that IP-10 plays a crucial role during the early immunization phase in the induction of immunity against neuroblastoma by scIL-12 gene therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocinas CXC/fisiología , Terapia Genética/métodos , Interleucina-12/genética , Neuroblastoma/inmunología , Neuroblastoma/prevención & control , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/biosíntesis , Inhibidores de la Angiogénesis/genética , Animales , Quimiocina CXCL10 , Quimiocinas CXC/antagonistas & inhibidores , Epítopos de Linfocito T/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Inyecciones Subcutáneas , Interleucina-12/administración & dosificación , Interleucina-12/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos A , Ratones SCID , Neuroblastoma/irrigación sanguínea , Neuroblastoma/metabolismo , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/trasplante , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
11.
Clin Cancer Res ; 7(3 Suppl): 856s-864s, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11300483

RESUMEN

Peripheral T-cell tolerance toward human carcinoembryonic self-antigen (CEA) was broken in CEA-transgenic C57BL/6J mice by an oral CEA-based DNA vaccine. This vaccine, delivered by the live, attenuated AroA- strain of Salmonella typhimurium (SL7207), induced tumor-protective immunity mediated by MHC class I-restricted CD8+ T cells. Activation of these T cells was indicated by increased secretion of proinflammatory cytokines IFN-gamma, interleukin (IL)-12 and granulocyte/macrophage-colony stimulating factor, as well as specific tumor rejection and growth suppression in vaccinated CEA-transgenic mice after a lethal challenge with murine MC38 colon carcinoma cells. These tumor cells were double transfected with CEA and the human epithelial cell adhesion molecule (Ep-CAM)/KSA and consequently served as a docking site for a recombinant antibody-IL2 fusion protein (KS1/4-IL2) recognizing KSA. Importantly, the efficacy of the tumor-protective immune response was markedly increased by boosts with this antibody-IL2 fusion protein, resulting in more effective tumor rejection coupled with increased expression of costimulatory molecules B7.2/B7.2 and intercellular adhesion molecule 1 (ICAM-1) on dendritic cells and intensified release of proinflammatory cytokines IFN-gamma, IL-12, and granulocyte/macrophage-colony stimulating factor from T cells of successfully vaccinated CEA-transgenic C57BL/6J mice. Increased T-cell activation mediated by boosts with KS1/4-IL2 fusion protein after tumor cell challenge was further indicated by expanded expression of T-cell activation markers CD25, CD28, CD69, and LFA-1. The application of such CEA-based DNA vaccines and its further improved versions may ultimately prove useful in combination therapies directed against human carcinomas expressing CEA self-antigens.


Asunto(s)
Vacunas contra el Cáncer , Antígeno Carcinoembrionario/metabolismo , Vacunas de ADN , Animales , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias del Colon/metabolismo , Citocinas/metabolismo , Molécula de Adhesión Celular Epitelial , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Immunoblotting , Molécula 1 de Adhesión Intercelular/metabolismo , Interferón gamma/metabolismo , Interleucina-12/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plásmidos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba
12.
Eur J Immunol ; 31(1): 250-8, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11265641

RESUMEN

Immunity to tumors relies on recirculating antigen-specific T cells. Whilst induction of antigen-specific T cells by immunotherapy has been convincingly proven, direct evidence for recirculation of such cells is still lacking. Here, employing a recently established in situ immunotherapy model for murine melanoma we directly demonstrate the redistribution of clonally expanded T cells. In this model IL-2 is targeted to the tumor microenvironment by means of specific antibody-IL-2 fusion proteins resulting in the expansion of T cells. The therapeutic effect of the fusion protein is not restricted to tumors expressing the targeted antigen, but extends to antigen negative variants of the tumor if present in the same animal. Analysis of the T cell infiltrate by quantitative reverse transcription-PCR revealed the presence of highly expressed TCR BV regions in both tumor variants. TCR clonotype mapping revealed that the high expressions of these regions were caused by clonal expansions and, notably, that these specific clonotypic TCR transcripts were identical in both tumors. Thus, T cell clones activated locally by targeted IL-2 therapy recirculate and mediate eradication of distant tumor sites not subjected to in situ cytokine therapy.


Asunto(s)
Anticuerpos/uso terapéutico , Interleucina-2/uso terapéutico , Melanoma Experimental/terapia , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Humanos , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
Immunity ; 14(2): 111-21, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11239444

RESUMEN

A recombinant antibody-lymphotoxin-alpha fusion protein induced an adaptive immune response protecting mice from melanoma. Importantly, this fusion protein elicited the formation of a lymphoid-like tissue in the tumor microenvironment containing L-selectin+ T cells and MHC class II+ antigen-presenting cells, as well as B and T cell aggregates. Furthermore, PNAd+/TCA4+ high endothelial venules were observed within the tumor, suggesting entry channels for naive T cell infiltrates. Over the course of therapy, a marked clonal expansion of certain TCR specificities occurred among tumor-infiltrating lymphocytes that displayed reactivity against melanoma cells and the TRP-2(180-188) peptide. Consequently, naive T cells may have been recruited to as well as primed and expanded in the lymphoid-like tissue induced by the lymphotoxin-alpha fusion protein at the tumor site.


Asunto(s)
Inmunotoxinas/uso terapéutico , Linfotoxina-alfa/uso terapéutico , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Endotelio Linfático/inmunología , Endotelio Linfático/patología , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica , Trasplante de Neoplasias , Neoplasias de Tejido Conjuntivo/inmunología , Neoplasias de Tejido Conjuntivo/patología , Neoplasias de Tejido Conjuntivo/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T/inmunología , Linfocitos T/patología , Trasplante Isogénico
14.
Cancer Res ; 61(4): 1500-7, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245457

RESUMEN

The huKS1/4-IL2 fusion protein, directed against the human epithelial cell adhesion molecule (huEpCAM) has been shown to induce a strong CD8+ T-cell-dependent, natural killer (NK) cell-independent, antitumor response in mice bearing the huEp-CAM-transfected CT26 colon cancer CT26-EpCAM. Here we investigate the effectiveness of huKS1/4-IL2 against CT26-Ep21.6, a subclone of CT26-EpCAM, expressing low levels of MHC class I. In vitro antibody-dependent cellular cytotoxicity (ADCC) assays in the presence of huKS1/4-IL2 demonstrate that murine NK cells from spleen and blood can kill CT26-Ep21.6 significantly better than they kill CT26-EpCAM. NK-mediated ADCC of CT26-EpCAM can be enhanced by blocking the murine NK cell-inhibitory receptor, Ly-49C. A potent in vivo antitumor effect was observed when BALB/c mice bearing experimental metastases of CT26-Ep21.6 were treated with huKS1/4-IL2. The depletion of NK cells during huKS1/4-IL2 treatment significantly reduced the antitumor effect against CT26-Ep21.6. Together our in vitro and in vivo data in the huEp-CAM-transfected CT26 models indicate that the amount of MHC class I expressed on the tumor target cell plays a critical role in the in vivo antitumor mechanism of huKS1/4-IL2 immunotherapy. A low MHC class I level favors NK cells as effectors, whereas a high level of MHC class I favors T cells as effectors. Given the heterogeneity of MHC class I expression seen in human tumors and the prevailing T-cell suppression in many cancer patients, the observation that huKS1/4-IL2 has the potential to effectively activate an NK cell-based antitumor response may be of potential clinical relevance.


Asunto(s)
Adenocarcinoma/inmunología , Antígenos H-2/inmunología , Inmunoconjugados/inmunología , Interleucina-2/inmunología , Neoplasias Pulmonares/inmunología , Adenocarcinoma/secundario , Adenocarcinoma/terapia , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Antineoplásicos/biosíntesis , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígenos de Neoplasias/inmunología , Moléculas de Adhesión Celular/inmunología , Relación Dosis-Respuesta Inmunológica , Molécula de Adhesión Celular Epitelial , Femenino , Antígenos H-2/biosíntesis , Inmunoconjugados/farmacología , Inmunoterapia/métodos , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/farmacología , Bazo/citología , Bazo/inmunología , Células Tumorales Cultivadas , Regulación hacia Arriba
15.
J Clin Oncol ; 18(24): 4077-85, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11118469

RESUMEN

PURPOSE: Ganglioside G(D2) is strongly expressed on the surface of human neuroblastoma cells. It has been shown that the chimeric human/murine anti-G(D2) monoclonal antibody (ch14.18) can induce lysis of neuroblastoma cells by antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. The purposes of the study were (1) to determine the maximum-tolerated dose (MTD) of ch14.18 in combination with standard dose granulocyte-macrophage colony-stimulating factor (GM-CSF) for patients with neuroblastoma who recently completed hematopoietic stem-cell transplantation (HSCT), and (2) to determine the toxicities of ch14.18 with GM-CSF in this setting. PATIENTS AND METHODS: Patients became eligible when the total absolute phagocyte count (APC) was greater than 1, 000/microL after HSCT. ch14.18 was infused intravenously over 5 hours daily for 4 consecutive days. Patients received GM-CSF 250 microg/m(2)/d starting at least 3 days before ch14.18 and continued for 3 days after the completion of ch14.18. The ch14.18 dose levels were 20, 30, 40, and 50 mg/m(2)/d. In the absence of progressive disease, patients were allowed to receive up to six 4-day courses of ch14.18 therapy with GM-CSF. Nineteen patients with neuroblastoma were treated. RESULTS: A total of 79 courses were administered. No toxic deaths occurred. The main toxicities were severe neuropathic pain, fever, nausea/vomiting, urticaria, hypotension, mild to moderate capillary leak syndrome, and neurotoxicity. Three dose-limiting toxicities were observed among six patients at 50 mg/m(2)/d: intractable neuropathic pain, grade 3 recurrent urticaria, and grade 4 vomiting. Human antichimeric antibody developed in 28% of patients. CONCLUSION: ch14.18 can be administered with GM-CSF after HSCT in patients with neuroblastoma with manageable toxicities. The MTD is 40 mg/m(2)/d for 4 days when given in this schedule with GM-CSF.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Gangliósidos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Trasplante de Células Madre Hematopoyéticas , Neuroblastoma/terapia , Adolescente , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Formación de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Niño , Preescolar , Terapia Combinada , Progresión de la Enfermedad , Relación Dosis-Respuesta Inmunológica , Esquema de Medicación , Femenino , Estudios de Seguimiento , Humanos , Masculino , Ratones , Neuroblastoma/inmunología , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/inmunología
16.
Med Pediatr Oncol ; 35(6): 641-6, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11107137

RESUMEN

BACKGROUND: The disruption of self-tolerance against neuroblastoma is the ultimate goal of an effective DNA-vaccine. PROCEDURE: Here we demonstrate the induction of a protective immunity against syngeneic murine NXS2 neuroblastoma in A/J mice, following vaccination with tyrosine hydroxylase (TH) derived antigens. Oral gene delivery was accomplished using an attenuated strain of Salmonella typhimurium as a carrier harboring vectors encoding for mTH antigens. RESULTS: Vaccination was effective in protecting animals from a lethal challenge with wild-type NXS2 tumor cells. CONCLUSIONS: These results provide the first evidence of the TH self antigen being recognized by T-cells and demonstrate that a TH-based DNA vaccine is a potentially useful immunotherapeutic strategy for neuroblastoma.


Asunto(s)
Neuroblastoma/prevención & control , Tirosina 3-Monooxigenasa , Vacunas de ADN , Animales , Técnicas de Transferencia de Gen , Ratones , Neuroblastoma/inmunología , Plásmidos , Tirosina 3-Monooxigenasa/genética
17.
Proc Natl Acad Sci U S A ; 97(21): 11500-4, 2000 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-11016973

RESUMEN

A major goal of cancer immunotherapy is the induction of a cell-mediated antitumor response in poorly immunogenic malignancies. We tested the hypothesis that this can be achieved by cytokine gene therapy with a novel histone H2A-based transient transfection procedure. This was tested by using cytokine genes encoding for IL-2 and a single chain IL-12 (scIL-12) fusion protein in a recently developed murine neuroblastoma model. Here, we demonstrate that cytokine gene transfer of IL-2 and scIL-12 with histone H2A results in the induction of an antitumor immune response that is superior in some respects to gene transfer with Superfect, a commercially available activated dendrimer commonly used to effect transfection with plasmids. Three lines of evidence support this contention. First, histone H2A-mediated transfection of IL-2 induces a natural killer cell-induced rejection of primary tumors in contrast to Superfect, which produces only a partial reduction in primary tumor growth. Second, the induction of a T cell-mediated protective tumor immunity following gene transfer of scIL-12 is more efficient with the histone H2A-mediated gene transfer because rejection of a lethal wild-type tumor cell challenge is accompanied by the greatest degree of MHC class I-restricted tumor cell killing in vitro. Third, histone H2A-mediated scIL-12 gene therapy induces the greatest release of mIFN-gamma from splenocytes of vaccinated animals in contrast to Superfect and other controls.


Asunto(s)
Histonas/genética , Interleucina-2/genética , Neuroblastoma/terapia , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Terapia Genética , Interferón gamma/metabolismo , Ratones , Neuroblastoma/genética , Neuroblastoma/inmunología , Neuroblastoma/patología , Linfocitos T Citotóxicos/inmunología , Transfección , Células Tumorales Cultivadas
19.
J Clin Invest ; 105(11): 1623-30, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10841521

RESUMEN

The induction of tumor-protective immunity against malignancies remains a major challenge in cancer immunotherapy. A novel, humanized anti-ganglioside-GD(2)-IL-2 immunocytokine (hu14.18-IL-2) induced CD8(+) T cells to eradicate established pulmonary metastases of B78-D14 murine melanoma, in a process that required help by CD4(+) T cells and was mediated by the CD40/CD40 ligand (CD40L) interaction. The anti-tumor effect was diminished in mice deficient in CD4(+) T-cells. Three lines of evidence show that CD4(+) T-cell help was mediated by CD40/CD40L interaction but not by endogenous IL-2 production. First, the hu14.18-IL-2-induced anti-tumor response is partially abrogated in C57BL/6J CD40L knockout (KO) mice in contrast to C57BL/6J IL-2 KO animals, in which the immunocytokine was completely effective. Second, partial abrogation of the anti-tumor effect is induced with anti-CD40L antibodies to the same extent as with CD4(+) T-cell depletion. Third, a complete anti-tumor response induced by hu14.18-IL-2 can be reconstituted in C57BL/6J CD40L KO mice by simultaneous stimulation with an anti-CD40 mAb. These results suggest that help provided by CD4(+) T cells via CD40/CD40L interactions in our tumor model is crucial for effective immunotherapy with an IL-2 immunocytokine.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/fisiología , Interleucina-2/uso terapéutico , Melanoma Experimental/terapia , Glicoproteínas de Membrana/fisiología , Animales , Células Presentadoras de Antígenos/fisiología , Antígenos CD40/genética , Ligando de CD40 , Femenino , Activación de Linfocitos , Melanoma Experimental/inmunología , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
20.
Immunol Res ; 21(2-3): 279-88, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10852128

RESUMEN

Targeting of cytokines into the tumor microenvironment using antibody-cytokine fusion proteins, called immunocytokines, represents a novel approach in cancer immunotherapy. This article summarizes therapeutic efficacy and immune mechanisms involved in targeting interleukin-2 (IL-2) to neuroectodermal tumors using ganglioside GD2-specific antibody-IL-2 fusion protein (ch14.18-IL-2). Treatment of established melanoma metastases with ch14.18-IL-2 resulted in eradication of disease followed by a vaccination effect protecting mice from lethal challenges with wild-type tumor calls. In a syngeneic neuroblastoma model, targeted IL-2 was effective in the amplification of a weak memory immune response previously induced by IL-12 gene therapy using an engineered linear version of this heterodimeric cytokine. These findings show that targeted IL-2 may provide an effective tool in cancer immunotherapy and establish the missing link between T cell-mediated vaccination and objective clinical responses.


Asunto(s)
Inmunoterapia , Interleucina-2/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Terapia Genética , Interleucina-2/genética , Ratones , Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...