Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Clin Cancer Res ; 24(1): 189-196, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28972044

RESUMEN

Purpose: In 2010, a Children's Oncology Group (COG) phase III randomized trial for patients with high-risk neuroblastoma (ANBL0032) demonstrated improved event-free survival (EFS) and overall survival (OS) following treatment with an immunotherapy regimen of dinutuximab, GM-CSF, IL2, and isotretinoin compared with treatment with isotretinoin alone. Dinutuximab, a chimeric anti-GD2 monoclonal antibody, acts in part via natural killer (NK) cells. Killer immunoglobulin-like receptors (KIR) on NK cells and their interactions with KIR-ligands can influence NK cell function. We investigated whether KIR/KIR-ligand genotypes were associated with EFS or OS in this trial.Experimental Design: We genotyped patients from COG study ANBL0032 and evaluated the effect of KIR/KIR-ligand genotypes on clinical outcomes. Cox regression models and log-rank tests were used to evaluate associations of EFS and OS with KIR/KIR-ligand genotypes.Results: In this trial, patients with the "all KIR-ligands present" genotype as well as patients with inhibitory KIR2DL2 with its ligand (HLA-C1) together with inhibitory KIR3DL1 with its ligand (HLA-Bw4) were associated with improved outcome if they received immunotherapy. In contrast, for patients with the complementary KIR/KIR-ligand genotypes, clinical outcome was not significantly different for patients who received immunotherapy versus those receiving isotretinoin alone.Conclusions: These data show that administration of immunotherapy is associated with improved outcome for neuroblastoma patients with certain KIR/KIR-ligand genotypes, although this was not seen for patients with other KIR/KIR-ligand genotypes. Further investigation of KIR/KIR-ligand genotypes may clarify their role in cancer immunotherapy and may enable KIR/KIR-ligand genotyping to be used prospectively for identifying patients likely to benefit from certain cancer immunotherapy regimens. Clin Cancer Res; 24(1); 189-96. ©2017 AACRSee related commentary by Cheung and Hsu, p. 3.


Asunto(s)
Genotipo , Neuroblastoma/genética , Neuroblastoma/mortalidad , Receptores KIR/genética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Ensayos Clínicos Fase III como Asunto , Femenino , Humanos , Inmunoterapia , Ligandos , Masculino , Neuroblastoma/inmunología , Neuroblastoma/terapia , Receptores KIR2DL1/genética , Receptores KIR2DL1/metabolismo , Receptores KIR3DL1/genética , Receptores KIR3DL1/metabolismo
2.
Carcinogenesis ; 35(3): 613-23, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24233838

RESUMEN

Previous studies have implicated cancer stem cells in tumor recurrence and revealed that the stem cell gene SOX2 plays an important role in the tumor cell resistance to apoptosis. Nonetheless, the mechanism by which SOX2 regulates apoptosis signals remained undefined. Here, we demonstrated the surprising finding that silencing of the SOX2 gene effectively induces apoptosis via the activation of death receptor and mitochondrial signaling pathways in human non-small cell lung cancer cells. Unexpectedly, reverse transcription-PCR analysis suggested that downregulation of SOX2 leads to activation of MAP4K4, previously implicated in cell survival. Evaluation of the apoptotic pathways revealed an increased expression of key inducers of apoptosis, including tumor necrosis factor-α and p53, with concurrent attenuation of Survivin. Although p53 appeared dispensable for this pathway, the loss of Survivin in SOX2-deficient cells appeared critical for the observed MAP4K4 induced cell death. Rescue experiments revealed that SOX2-silencing-mediated killing was blocked by ectopic expression of Survivin, or by reduction of MAP4K4 expression. Clinically, expressions of Survivin and SOX2 were highly correlated with each other. The results reveal a key target of SOX2 expression and highlight the unexpected context-dependent role for MAP4K4, a pluripotent activator of several mitogen-activated protein kinase pathways, in regulating tumor cell survival.


Asunto(s)
Apoptosis/fisiología , Proteínas Inhibidoras de la Apoptosis/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Silenciador del Gen , Humanos , Ratones , Ratones SCID , Factores de Transcripción SOXB1/genética , Survivin
3.
PLoS One ; 8(8): e73607, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24014113

RESUMEN

Although early detection of breast cancer improved in recent years, prognosis of patients with late stage breast cancer remains poor, mostly due to development of multidrug resistance (MDR) followed by tumor recurrence. Cancer stem cells (CSCs), with higher drug efflux capability and other stem cell-like properties, are concentrated in a side population (SP) of cells, which were proposed to be responsible for MDR and tumor repopulation that cause patients to succumb to breast cancer. Therefore, targeting of CSCs as an adjuvant to chemotherapy should be able to provide a more effective treatment of this disease. Here, we used IMD-0354, an inhibitor of NF-κB, identified for targeting CSCs, in a combination therapy with doxorubicin encapsulated in targeted nanoparticles. IMD-0354 did target CSCs, evidenced by a decrease in the SP, demonstrated by the inhibition of the following: dye/drug efflux, reduction in ABC transporters as well as in colony formation in soft agar and low attachment plates. Decrease of stem-like gene expression of Oct4, Nanog and Sox2, and apoptosis resistance related to the Survivin gene also was observed after treatment with this compound. In addition, IMD-0354 targeted non-CSCs as indicated by reducing viability and increasing apoptosis. Targeted drug delivery, achieved with a legumain inhibitor, proved to enhance drug delivery under hypoxia, a hallmark of the tumor microenvironment, but not under normoxia. Together, this allowed a safe, non-toxic delivery of both anticancer agents to the tumor microenvironment of mice bearing syngeneic metastatic breast cancer. Targeting both bulk tumor cells with a chemotherapeutic agent and CSCs with IMD-0354 should be able to reduce MDR. This could eventually result in decreasing tumor recurrences and/or improve the outcome of metastatic disease.


Asunto(s)
Benzamidas/farmacología , Quimioradioterapia Adyuvante , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Mamarias Animales/terapia , Células Madre Neoplásicas/metabolismo , Animales , Antibióticos Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Doxorrubicina/farmacología , Sistemas de Liberación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/patología
4.
Cancer Biol Ther ; 14(6): 511-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23760493

RESUMEN

The yes-associated protein (YAP) transcription co-activator has been reported either as an oncogene candidate or a tumor suppressor. Liver tissue chips revealed that about 51.4% human hepatocellular carcinoma (HCC) samples express YAP and 32.9% HCC samples express phosphorylated YAP. In this study, we found that chemotherapy increased YAP protein expression and nuclear translocation in HepG2 cells, as well as p53 protein expression and nuclear translocation. However, little is known about YAP functions during chemotherapy. Our results show that overexpression of YAP increases chemosensitivity of HepG2 cells during chemotherapy. Dominant negative transfection of Flag-S94A (TEAD binding domain mutant) or Flag-W1W2 (WW domain mutant) to HepG2 cells decreases p53 expression/ nuclear translocation and chemosensitivity when compared with control HepG2 cells. Furthermore, rescue transfection of Flag-5SA-S94A or Flag-5SA-W1W2, respectively to HepG2 cells regains p53 expression/nuclear translocation and chemosensitivity. These results indicate that YAP promotes chemosensitivity by modulating p53 during chemotherapy and both TEAD and WW binding domains are required for YAP-mediated p53 function. ChIP assay results also indicated that YAP binds directly to the p53 promoter to improve its expression. In addition, p53 could positively feedback YAP expression through binding to the YAP promoter. Taken together, our current data indicate that YAP functions as a tumor suppressor that enhances apoptosis by modulating p53 during chemotherapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Carcinoma Hepatocelular/metabolismo , Resistencia a Antineoplásicos , Neoplasias Hepáticas/metabolismo , Fosfoproteínas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Antibióticos Antineoplásicos/farmacología , Apoptosis , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/farmacología , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Hígado/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción , Proteína p53 Supresora de Tumor/genética , Proteínas Señalizadoras YAP
5.
Cancer Immunol Immunother ; 62(8): 1303-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23661160

RESUMEN

Hu14.18-IL2 is an immunocytokine (IC) consisting of human IL-2 linked to hu14.18 mAb, which recognizes GD2 disialoganglioside. Phase II clinical trials of intravenous-hu14.18-IL2 (IV-IC) in neuroblastoma and melanoma are underway, and have already demonstrated activity in neuroblastoma. In our Phase II trial, lower neuroblastoma burden at the time of treatment was associated with a greater likelihood of clinical response to IV-IC. We have previously shown that intratumoral-hu14.18-IL2 (IT-IC) compared to IV-IC results in enhanced local and systemic antitumor activity in tumor-bearing mice. We utilized a mouse model to investigate the impact of tumor burden on hu14.18-IL2 treatment efficacy in IV- versus IT-treated animals. Studies presented here describe the analyses of tumor burden at the initiation of treatment and its effects on treatment efficacy, survival, and tumor-infiltrating leukocytes in A/J mice bearing subcutaneous NXS2 neuroblastoma. We show that smaller tumor burden at treatment initiation is associated with increased infiltration of NK and CD8+ T cells and increased overall survival. NXS2 tumor shrinkage shortly after completion of the 3 days of hu14.18-IL2 treatment is necessary for long-term survival. This model demonstrates that tumor size is a strong predictor of hu14.18-IL2-induced lymphocyte infiltration and treatment outcome.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Linfocitos T CD8-positivos/inmunología , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Neuroblastoma/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/genética , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Interleucina-2/administración & dosificación , Interleucina-2/genética , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Neuroblastoma/patología , Neuroblastoma/terapia , Pronóstico , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/inmunología
6.
Crit Rev Oncog ; 18(1-2): 115-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23237555

RESUMEN

Cancer immunotherapy is in the midst of a major paradigm shift from an approach primarily focused on attacking tumor cells to a strategy also targeting the tumor microenvironment (TME). This strategy is designed for the use of combination therapies, several of which are reviewed here. Particular emphasis is placed on targeting such components of the TME as tumor-associated macrophages (TAMs) and cancer-associated fibroblasts (CAFs) overexpressing specific therapy targets such as Legumain, an asparaginyl endopeptidase, proto-oncogene Fra-1, transcription factor Stat3 and fibroblast activation protein (FAP) as well as HER-2, respectively. The use of DNA vaccines directed against some of these targets overexpressed on both breast tumor cells as well as TAMs and CAFs in the TME results in the elimination of tumor growth, progression, metastasis and recurrence in mouse tumor models. This type of cancer therapy is significantly improved in efficacy by a strategy specifically designed to combine immunotherapies, including DNA vaccines, with a novel chemotherapy featuring highly effective nanoparticle-mediated drug delivery, specifically targeted to tumor cells as well as key components of the TME, leading to its modulation and subsequent elimination of tumor growth, metastasis, and, most importantly, suppression of tumor recurrence.


Asunto(s)
Neoplasias de la Mama , Terapia Molecular Dirigida , Microambiente Tumoral , Vacunas de ADN/administración & dosificación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Cisteína Endopeptidasas/administración & dosificación , Femenino , Fibroblastos/inmunología , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Macrófagos/inmunología , Macrófagos/metabolismo , Proto-Oncogenes Mas , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
7.
Stem Cells ; 31(2): 248-58, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169551

RESUMEN

The cancer stem cell (CSC) hypothesis has gained significant recognition as a descriptor of tumorigenesis. Additionally, tumor-associated macrophages (TAMs) are known to promote growth and metastasis of breast cancer. However, it is not known whether TAMs mediate tumorigenesis through regulation of breast CSCs. Here, we report that TAMs promote CSC-like phenotypes in murine breast cancer cells by upregulating their expression of Sox-2. These CSC-like phenotypes were characterized by increased Sox-2, Oct-4, Nanog, AbcG2, and Sca-1 gene expression, in addition to increased drug-efflux capacity, resistance to chemotherapy, and increased tumorigenicity in vivo. Downregulation of Sox-2 in tumor cells by siRNA blocked the ability of TAMs to induce these CSC-like phenotypes and inhibited tumor growth in vivo. Furthermore, we identified a novel epidermal growth factor receptor (EGFR)/signal transducers and activators of transcription 3 (Stat3)/Sox-2 paracrine signaling pathway between macrophages and mouse breast cancer cells that is required for macrophage-induced upregulation of Sox-2 and CSC phenotypes in tumor cells. We showed that this crosstalk was effectively blocked by the small molecule inhibitors AG1478 or CDDO-Im against EGFR and Stat3, respectively. Therefore, our report identifies a novel role for TAMs in breast CSC regulation and establishes a rationale for targeting the EGFR/Stat3/Sox-2 signaling pathway for CSC therapy.


Asunto(s)
Receptores ErbB/genética , Regulación Neoplásica de la Expresión Génica , Macrófagos/metabolismo , Neoplasias Mamarias Animales/genética , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/genética , Factor de Transcripción STAT3/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Apoptosis/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Imidazoles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteína Homeótica Nanog , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/farmacología , Quinazolinas/farmacología , ARN Interferente Pequeño/genética , Factores de Transcripción SOXB1/antagonistas & inhibidores , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Tirfostinos/farmacología
8.
J Immunol ; 189(5): 2656-64, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22844125

RESUMEN

hu14.18-IL-2 (IC) is an immunocytokine consisting of human IL-2 linked to hu14.18 mAb, which recognizes the GD2 disialoganglioside. Phase 2 clinical trials of i.v. hu14.18-IL-2 (i.v.-IC) in neuroblastoma and melanoma are underway and have already demonstrated activity in neuroblastoma. We showed previously that intratumoral hu14.18-IL-2 (IT-IC) results in enhanced antitumor activity in mouse models compared with i.v.-IC. The studies presented in this article were designed to determine the mechanisms involved in this enhanced activity and to support the future clinical testing of intratumoral administration of immunocytokines. Improved survival and inhibition of growth of both local and distant tumors were observed in A/J mice bearing s.c. NXS2 neuroblastomas treated with IT-IC compared with those treated with i.v.-IC or control mice. The local and systemic antitumor effects of IT-IC were inhibited by depletion of NK cells or T cells. IT-IC resulted in increased NKG2D receptors on intratumoral NKG2A/C/E⁺ NKp46⁺ NK cells and NKG2A/C/E⁺ CD8⁺ T cells compared with control mice or mice treated with i.v.-IC. NKG2D levels were augmented more in tumor-infiltrating lymphocytes compared with splenocytes, supporting the localized nature of the intratumoral changes induced by IT-IC treatment. Prolonged retention of IC at the tumor site was seen with IT-IC compared with i.v.-IC. Overall, IT-IC resulted in increased numbers of activated T and NK cells within tumors, better IC retention in the tumor, enhanced inhibition of tumor growth, and improved survival compared with i.v.-IC.


Asunto(s)
Antineoplásicos/uso terapéutico , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neuroblastoma/inmunología , Neuroblastoma/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Inyecciones Intralesiones , Interleucina-2/administración & dosificación , Interleucina-2/metabolismo , Interleucina-2/uso terapéutico , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos A , Ratones Endogámicos C57BL , Neuroblastoma/patología , Distribución Aleatoria , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
9.
PLoS One ; 7(5): e36326, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615765

RESUMEN

Recent studies demonstrated that cancer stem cells (CSCs) have higher tumorigenesis properties than those of differentiated cancer cells and that transcriptional factor-SOX2 plays a vital role in maintaining the unique properties of CSCs; however, the function and underlying mechanism of SOX2 in carcinogenesis of lung cancer are still elusive. This study applied immunohistochemistry to analyze the expression of SOX2 in human lung tissues of normal individuals as well as patients with adenocarcinoma, squamous cell carcinoma, and large cell and small cell carcinoma and demonstrated specific overexpression of SOX2 in all types of lung cancer tissues. This finding supports the notion that SOX2 contributes to the tumorigenesis of lung cancer cells and can be used as a diagnostic probe. In addition, obviously higher expression of oncogenes c-MYC, WNT1, WNT2, and NOTCH1 was detected in side population (SP) cells than in non-side population (NSP) cells of human lung adenocarcinoma cell line-A549, revealing a possible mechanism for the tenacious tumorigenic potential of CSCs. To further elucidate the function of SOX2 in tumorigenesis of cancer cells, A549 cells were established with expression of luciferase and doxycycline-inducible shRNA targeting SOX2. We found silencing of SOX2 gene reduces the tumorigenic property of A549 cells with attenuated expression of c-MYC, WNT1, WNT2, and NOTCH1 in xenografted NOD/SCID mice. By using the RNA-Seq method, an additional 246 target cancer genes of SOX2 were revealed. These results present evidence that SOX2 may regulate the expression of oncogenes in CSCs to promote the development of human lung cancer.


Asunto(s)
Redes Reguladoras de Genes , Neoplasias Pulmonares/genética , Oncogenes , Factores de Transcripción SOXB1/genética , Transcripción Genética , Secuencia de Bases , Línea Celular Tumoral , Transformación Celular Neoplásica , Cartilla de ADN , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/patología , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
Cancer Res ; 72(14): 3451-6, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22586064

RESUMEN

Fra-1 is a member of the Fos transcription factor family that is highly expressed in multiple cancers, playing important roles in transformation, proliferation, and metastasis. In this study, we observed an inverse correlation between the expression of Fra-1 in human stage II breast cancer tissues and the corresponding level of clinical chemoresistance. Extending these findings in vitro, we found that knockdown of Fra-1 in breast tumor cells was sufficient to confer resistance to doxorubicin and cyclophosphamide, whereas enhanced Fra-1 expression could render these cells chemosensitive. The tumor cell side population, which is enriched for cancer stem cells, was found to be associated with chemoresistance. Increased side population fractions were detected among tumor cell lines subjected to Fra-1 knockdown. In contrast, enhanced expression of Fra-1 was correlated with a decreased side population fraction, and significantly, this finding was recapitulated in vivo, where tumors with enhanced expression of Fra-1 were found to have blunted growth. Tumor cells subjected to Fra-1 knockdown grew faster and were larger in size. Taken together, our findings suggest that Fra-1 may be an important prognostic marker for breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Resistencia a Antineoplásicos , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-fos/fisiología , Animales , Antineoplásicos/farmacología , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/patología , Línea Celular Tumoral , Ciclofosfamida/farmacología , Progresión de la Enfermedad , Doxorrubicina/farmacología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/patología , Pronóstico , Proteínas Proto-Oncogénicas c-fos/genética
11.
Int Immunopharmacol ; 11(11): 1877-86, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21854876

RESUMEN

We evaluated the anti-tumor effect of Resveratrol (RV) on M21 and NXS2 tumor cell lines and its immunosuppressive activity on human and murine immune cells to determine the potential for combining RV and immunotherapy. In vitro, concentrations of RV≥25 mcM, inhibited cell proliferation, blocked DNA synthesis and induced G1 phase arrest in tumor and immune cells. RV at 12-50 mcM inhibited antibody dependent cell mediated cytotoxicity (ADCC) of tumor cells facilitated by the hu14.18-IL2 immunocytokine (IC). The in vivo anti-tumor and immunomodulating activity of RV given systemically were assessed in mice. Results showed that this RV regimen inhibited the growth of NXS2 tumors in vivo but did not appear to interfere with blood cell count, splenocyte or macrophage function. Thus, RV may be a candidate for combining with immunotherapy.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Antineoplásicos Fitogénicos/farmacología , Inmunoterapia , Neoplasias/tratamiento farmacológico , Estilbenos/farmacología , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Femenino , Citometría de Flujo , Humanos , Inmunoterapia/métodos , Leucocitos Mononucleares/efectos de los fármacos , Ratones , Ratones Desnudos , Neoplasias/inmunología , Neoplasias/patología , Resveratrol , Bazo/citología , Bazo/efectos de los fármacos
12.
Cancer Res ; 71(17): 5688-96, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21784871

RESUMEN

The tumor microenvironment (TME) mediates immunosuppression resulting in tumor cell escape from immune surveillance and cancer vaccine failure. Immunosuppression is mediated by the STAT-3 transcription factor, which potentiates signaling in tumor and immune cells. Because immunosuppression continues to be a major inhibitor of cancer vaccine efficacy, we examined in this study whether therapeutically targeted delivery of a synthetic STAT-3 inhibitor to the TME, combined with an HER-2 DNA vaccine can improve immune surveillance against HER-2(+) breast cancer and prevent its recurrence. To this end, we developed a novel ligand-targeted nanoparticle (NP) encapsulating a CDDO-Im payload capable of specific delivery to the TME, which showed an effective therapeutic inhibition of STAT-3 activation in primary tumors. Furthermore, we showed that treatment with these NPs resulted in priming of the immune TME, characterized by increased IFN-γ, p-STAT-1, GM-CSF, IL-2, IL-15, and IL-12b and reduced TGF-ß, IL-6, and IL-10 protein expression. In addition, we found significantly increased tumor infiltration by activated CD8(+) T cells, M1 macrophages, and dendritic cells. These changes correlated with delayed growth of orthotopic 4TO7 breast tumors and, when combined with an HER-2 DNA vaccine, prevented HER-2(+) primary tumor recurrence in immunocompetent mice. Furthermore, antitumor T-cell responses were enhanced in splenocytes isolated from mice treated with this combination therapy. Together, these data show effective protection from cancer recurrence through improved immune surveillance against a tumor-specific antigen.


Asunto(s)
Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/uso terapéutico , Neoplasias Mamarias Experimentales/terapia , Recurrencia Local de Neoplasia/prevención & control , Receptor ErbB-2/antagonistas & inhibidores , Microambiente Tumoral , Vacunas de ADN/uso terapéutico , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Femenino , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos BALB C , Nanopartículas/administración & dosificación , Receptor ErbB-2/inmunología , Factor de Transcripción STAT3/antagonistas & inhibidores , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
13.
Nanomedicine ; 7(6): 665-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21419870

RESUMEN

Unresolved problems associated with ligand-targeting of liposomal nanoparticles (NPs) to solid tumors include variable target receptor expression due to genetic heterogeneity and insufficient target specificity, leading to systemic toxicities. This study addresses these issues by developing a novel ligand-targeting strategy for liposomal NPs using RR-11a, a synthetic enzyme inhibitor of Legumain, an asparaginyl endopeptidase. Cell-surface expression of Legumain is driven by hypoxic stress, a hallmark of solid tumors. Legumain-targeted RR-11a-coupled NPs revealed high ligand-receptor affinity, enhanced solid-tumor penetration and uptake by tumor cells. Treatment of tumor-bearing mice with RR-11a-coupled NPs encapsulating doxorubicin resulted in improved tumor selectivity and drug sensitivity, leading to complete inhibition of tumor growth. These antitumor effects were achieved while eliminating systemic drug toxicity. Therefore, synthetic enzyme inhibitors, such as RR-11a, represent a new class of compounds that can be used for highly specific ligand-targeting of NPs to solid tumors. FROM THE CLINICAL EDITOR: This study addresses the problems associated with ligand-targeting of liposomal nanoparticles to solid tumors with variable target receptor expression. A novel and efficacious targeting strategy has been developed towards a synthetic enzyme inhibitor of Legumain. The authors demonstrate successful tumor growth inhibiting effect while eliminating systemic drug toxicity in an animal model using this strategy.


Asunto(s)
Antineoplásicos/administración & dosificación , Cisteína Endopeptidasas/metabolismo , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liposomas/química , Nanopartículas/química , Inhibidores de Proteasas/metabolismo , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Cisteína Endopeptidasas/genética , Doxorrubicina/farmacocinética , Doxorrubicina/uso terapéutico , Femenino , Regulación Neoplásica de la Expresión Génica , Ligandos , Ratones , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/patología , Inhibidores de Proteasas/química
14.
Cancer Immunol Immunother ; 60(5): 731-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21340652

RESUMEN

We investigated the anti-tumor effect of peritumoral resveratrol in combination with immunotherapy in vivo in neuroblastoma-bearing mice. Subcutaneous NXS2 tumors were induced in A/J mice. On day 10, some mice received 15 mcg of intravenous immunocytokine for 5 days, mice received 20 mg of peritumoral resveratrol twice a week (starting on day 12) for a total of 5 injections, and a separate group received a combination of both regimens. Tumor progression and survival were assessed every 3-4 days. Blood and primary tumor tissue samples were collected on day 20 for Complete Blood Count and CD45 immunohistochemistry and histology, respectively. The primary tumor regressed in all mice receiving peritumoral resveratrol. Most of these mice receiving peritumoral resveratrol alone developed metastatic tumors and recurrence of the primary tumor after cessation of therapy. When resveratrol and immunocytokine regimens were combined, 61% of the mice receiving this combination therapy resolved their primary tumors and survived without developing metastatic tumors, compared to 15 and 13% receiving resveratrol or immunocytokine alone, respectively. None of the therapeutic regimes prevented lymphocyte infiltration or affected the complete blood count. Greater necrosis was observed microscopically in tumors from mice receiving the combination therapy. These results demonstrate that the combination therapy of peritumoral resveratrol plus intravenous immunocytokine provides better anti-tumor effects in this model than either therapy alone.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Inmunoglobulina G/uso terapéutico , Interleucina-2/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/terapia , Estilbenos/uso terapéutico , Animales , Antineoplásicos Fitogénicos/sangre , Línea Celular Tumoral , Terapia Combinada , Citometría de Flujo , Gangliósidos/análisis , Gangliósidos/sangre , Inmunoterapia , Antígenos Comunes de Leucocito , Leucocitos/efectos de los fármacos , Ratones , Ratones Endogámicos A , Recurrencia Local de Neoplasia , Neuroblastoma/inmunología , Resveratrol , Estilbenos/sangre , Sobrevida , Carga Tumoral/efectos de los fármacos
15.
J Leukoc Biol ; 89(4): 625-38, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21248148

RESUMEN

The hu14.18-IL2 (EMD 273063) IC, consisting of a GD(2)-specific mAb genetically engineered to two molecules of IL-2, is in clinical trials for treatment of GD(2)-expressing tumors. Anti-tumor activity of IC in vivo and in vitro involves NK cells. We studied the kinetics of retention of IC on the surface of human CD25(+)CD16(-) NK cell lines (NKL and RL12) and GD(2)(+) M21 melanoma after IC binding to the cells via IL-2R and GD(2), respectively. For NK cells, ∼ 50% of IC was internalized by 3 h and ∼ 90% by 24 h of cell culture. The decrease of surface IC levels on NK cells correlated with the loss of their ability to bind to tumor cells and mediate antibody-dependent cellular cytotoxicity in vitro. Unlike NK cells, M21 cells retained ∼ 70% of IC on the surface following 24 h of culture and maintained the ability to become conjugated and lysed by NK cells. When NKL cells were injected into M21-bearing SCID mice, IT delivery of IC augmented NK cell migration into the tumor. These studies demonstrate that once IC binds to the tumor, it is present on the tumor surface for a prolonged time, inducing the recruitment of NK cells to the tumor site, followed by tumor cell killing.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos , Interleucina-2/farmacología , Células Asesinas Naturales/inmunología , Melanoma/inmunología , Melanoma/terapia , Animales , Citometría de Flujo , Humanos , Inmunofenotipificación , Células Asesinas Naturales/efectos de los fármacos , Melanoma/genética , Ratones , Ratones SCID , Células Tumorales Cultivadas
16.
N Engl J Med ; 363(14): 1324-34, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20879881

RESUMEN

BACKGROUND: Preclinical and preliminary clinical data indicate that ch14.18, a monoclonal antibody against the tumor-associated disialoganglioside GD2, has activity against neuroblastoma and that such activity is enhanced when ch14.18 is combined with granulocyte-macrophage colony-stimulating factor (GM-CSF) or interleukin-2. We conducted a study to determine whether adding ch14.18, GM-CSF, and interleukin-2 to standard isotretinoin therapy after intensive multimodal therapy would improve outcomes in high-risk neuroblastoma. METHODS: Patients with high-risk neuroblastoma who had a response to induction therapy and stem-cell transplantation were randomly assigned, in a 1:1 ratio, to receive standard therapy (six cycles of isotretinoin) or immunotherapy (six cycles of isotretinoin and five concomitant cycles of ch14.18 in combination with alternating GM-CSF and interleukin-2). Event-free survival and overall survival were compared between the immunotherapy group and the standard-therapy group, on an intention-to-treat basis. RESULTS: A total of 226 eligible patients were randomly assigned to a treatment group. In the immunotherapy group, a total of 52% of patients had pain of grade 3, 4, or 5, and 23% and 25% of patients had capillary leak syndrome and hypersensitivity reactions, respectively. With 61% of the number of expected events observed, the study met the criteria for early stopping owing to efficacy. The median duration of follow-up was 2.1 years. Immunotherapy was superior to standard therapy with regard to rates of event-free survival (66±5% vs. 46±5% at 2 years, P=0.01) and overall survival (86±4% vs. 75±5% at 2 years, P=0.02 without adjustment for interim analyses). CONCLUSIONS: Immunotherapy with ch14.18, GM-CSF, and interleukin-2 was associated with a significantly improved outcome as compared with standard therapy in patients with high-risk neuroblastoma. (Funded by the National Institutes of Health and the Food and Drug Administration; ClinicalTrials.gov number, NCT00026312.)


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Gangliósidos/inmunología , Inmunoterapia , Neuroblastoma/terapia , Anticuerpos Monoclonales/efectos adversos , Antineoplásicos/uso terapéutico , Niño , Preescolar , Terapia Combinada , Quimioterapia Combinada , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunoterapia/efectos adversos , Lactante , Análisis de Intención de Tratar , Interleucina-2/uso terapéutico , Isotretinoína/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/mortalidad , Trasplante de Células Madre , Análisis de Supervivencia
17.
Cancer Res ; 70(23): 9554-61, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20935224

RESUMEN

Response to immunocytokine (IC) therapy is dependent on natural killer cells in murine neuroblastoma (NBL) models. Furthermore, killer immunoglobulin-like receptor (KIR)/KIR-ligand mismatch is associated with improved outcome to autologous stem cell transplant for NBL. Additionally, clinical antitumor response to monoclonal antibodies has been associated with specific polymorphic-FcγR alleles. Relapsed/refractory NBL patients received the hu14.18-IL2 IC (humanized anti-GD2 monoclonal antibody linked to human IL2) in a Children's Oncology Group phase II trial. In this report, these patients were genotyped for KIR, HLA, and FcR alleles to determine whether KIR receptor-ligand mismatch or specific FcγR alleles were associated with antitumor response. DNA samples were available for 38 of 39 patients enrolled: 24 were found to have autologous KIR/KIR-ligand mismatch; 14 were matched. Of the 24 mismatched patients, 7 experienced either complete response or improvement of their disease after IC therapy. There was no response or comparable improvement of disease in patients who were matched. Thus KIR/KIR-ligand mismatch was associated with response/improvement to IC (P = 0.03). There was a trend toward patients with the FcγR2A 131-H/H genotype showing a higher response rate than other FcγR2A genotypes (P = 0.06). These analyses indicate that response or improvement of relapsed/refractory NBL patients after IC treatment is associated with autologous KIR/KIR-ligand mismatch, consistent with a role for natural killer cells in this clinical response.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Interleucina-2/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Receptores de IgG/genética , Receptores KIR/genética , Adolescente , Niño , Preescolar , Genotipo , Antígenos HLA/genética , Antígenos HLA-B/genética , Antígenos HLA-C/genética , Humanos , Lactante , Células Asesinas Naturales/metabolismo , Ligandos , Neuroblastoma/genética , Polimorfismo Genético , Resultado del Tratamiento , Adulto Joven
18.
J Clin Oncol ; 28(33): 4969-75, 2010 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-20921469

RESUMEN

PURPOSE: The hu14.18-IL2 fusion protein consists of interleukin-2 molecularly linked to a humanized monoclonal antibody that recognizes the GD2 disialoganglioside expressed on neuroblastoma cells. This phase II study assessed the antitumor activity of hu14.18-IL2 in two strata of patients with recurrent or refractory neuroblastoma. PATIENTS AND METHODS: Hu14.18-IL2 was given intravenously (12 mg/m(2)/daily) for 3 days every 4 weeks for patients with disease measurable by standard radiographic criteria (stratum 1) and for patients with disease evaluable only by [(123)I]metaiodobenzylguanidine (MIBG) scintigraphy and/or bone marrow (BM) histology (stratum 2). Response was established by independent radiology review as well as BM histology and immunocytology, and durability was assessed by repeat evaluation after more than 3 weeks. RESULTS: Thirty-nine patients were enrolled (36 evaluable). No responses were seen in stratum 1 (n = 13). Of 23 evaluable patients in stratum 2, five patients (21.7%) responded; all had a complete response (CR) of 9, 13, 20, 30, and 35+ months duration. Grade 3 and 4 nonhematologic toxicities included capillary leak, hypoxia, pain, rash, allergic reaction, elevated transaminases, and hyperbilirubinemia. Two patients required dopamine for hypotension, and one patient required ventilatory support for hypoxia. Most toxicities were reversible within a few days of completing a treatment course and were expected based on phase I results. CONCLUSION: Patients with disease evaluable only by MIBG and/or BM histology had a 21.7% CR rate to hu14.8-IL2, whereas patients with bulky disease did not respond. Hu14.18-IL2 warrants further testing in children with nonbulky high-risk neuroblastoma.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Interleucina-2/uso terapéutico , Neuroblastoma/tratamiento farmacológico , Adolescente , Adulto , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales/inmunología , Niño , Preescolar , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Lactante , Interleucina-2/efectos adversos , Interleucina-2/sangre , Interleucina-2/inmunología , Recuento de Linfocitos , Neuroblastoma/mortalidad , Receptores de Interleucina-2/sangre
19.
Exp Dermatol ; 19(2): 157-64, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19849715

RESUMEN

Complex biological processes often require in vivo analysis, and many important research advances have been made using mice as a model for the study of various biological systems. Cutaneous melanomas are tumors originating from skin melanocytes, which are present in hair follicles, and interfollicular epidermal and dermal layers. Until recently, mouse melanoma models were largely based on transplantation models, i.e. transplantation of either syngeneic or xenogeneic melanoma cells into wild type or genetically modified animals. More recently, however, the use of novel technologies specifically modifying the genome allows for the generation of mouse strains, which may develop spontaneous melanoma. Nevertheless, it should be kept in mind that animal models provide only an approximation of reality in humans. In this review, we will discuss a representative selection of currently available transplantation and transgenic melanoma models; despite the fact that this selection will be biased by personal experience, we are confident to demonstrate how the use of mouse melanoma models facilitates translational research in several biomedical disciplines.


Asunto(s)
Modelos Animales de Enfermedad , Melanoma Experimental , Ratones , Animales , Humanos , Melanoma Experimental/genética , Trasplante Heterólogo , Trasplante Isogénico
20.
PLoS One ; 4(11): e7965, 2009 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-19956757

RESUMEN

BACKGROUND: Local inflammation associated with solid tumors commonly results from factors released by tumor cells and the tumor stroma, and promotes tumor progression. Cancer associated fibroblasts comprise a majority of the cells found in tumor stroma and are appealing targets for cancer therapy. Here, our aim was to determine the efficacy of targeting cancer associated fibroblasts for the treatment of metastatic breast cancer. METHODOLOGY/PRINCIPAL FINDINGS: We demonstrate that cancer associated fibroblasts are key modulators of immune polarization in the tumor microenvironment of a 4T1 murine model of metastatic breast cancer. Elimination of cancer associated fibroblasts in vivo by a DNA vaccine targeted to fibroblast activation protein results in a shift of the immune microenvironment from a Th2 to Th1 polarization. This shift is characterized by increased protein expression of IL-2 and IL-7, suppressed recruitment of tumor-associated macrophages, myeloid derived suppressor cells, T regulatory cells, and decreased tumor angiogenesis and lymphangiogenesis. Additionally, the vaccine improved anti-metastatic effects of doxorubicin chemotherapy and enhanced suppression of IL-6 and IL-4 protein expression while increasing recruitment of dendritic cells and CD8(+) T cells. Treatment with the combination therapy also reduced tumor-associated Vegf, Pdgfc, and GM-CSF mRNA and protein expression. CONCLUSIONS/SIGNIFICANCE: Our findings demonstrate that cancer associated fibroblasts promote tumor growth and metastasis through their role as key modulators of immune polarization in the tumor microenvironment and are valid targets for therapy of metastatic breast cancer.


Asunto(s)
Fibroblastos/citología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Fibroblastos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Interleucina-2/metabolismo , Interleucina-7/metabolismo , Linfocinas/metabolismo , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células TH1/citología , Células Th2/citología , Factor A de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA