Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 71(1): 153-164, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34043024

RESUMEN

Neuroblastoma (NBL) accounts for a disproportionate number of deaths among childhood malignancies despite intensive multimodal therapy that includes antibody targeting disialoganglioside GD2, a NBL antigen. Unfortunately, resistance to anti-GD2 immunotherapy is frequent and we aimed to investigate mechanisms of resistance in NBL. GD2 expression was quantified by flow cytometry and anti-GD2 antibody internalization was measured using real-time microscopy in 20 human NBL cell lines. Neutrophil-mediated antibody-dependent cellular cytotoxicity (ADCC) assays were performed on a subset of the cell lines (n = 12), and results were correlated with GD2 expression and antibody internalization. GD2 was expressed on 19 of 20 NBL cell lines at variable levels, and neutrophil-mediated ADCC was observed only in GD2-expressing cell lines. We found no correlation between level of GD2 expression and sensitivity to neutrophil-mediated ADCC, suggesting that GD2 expression of many cell lines was above a threshold required for maximal ADCC, such that expression level could not be used to predict subsequent cytotoxicity. Instead, anti-GD2 antibody internalization, a process that occurred universally but differentially across GD2-expressing NBL cell lines, was inversely correlated with ADCC. Treatment with endocytosis inhibitors EIPA, chlorpromazine, MBCD, and cytochalasin-D showed potential to inhibit antibody internalization; however, only MBCD resulted in significantly increased sensitivity to neutrophil-mediated ADCC in 4 of 4 cell lines in vitro. Our data suggest that antibody internalization may represent a novel mechanism of immunotherapy escape by NBL and provide proof-of-principle that targeting pathways involved in antibody internalization may improve the efficacy of anti-GD2 immunotherapies.


Asunto(s)
Anticuerpos/química , Resistencia a Medicamentos , Gangliósidos/química , Inmunoterapia/métodos , Neuroblastoma/inmunología , Neuroblastoma/terapia , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Línea Celular Tumoral , Endocitosis , Citometría de Flujo , Gangliósidos/inmunología , Humanos , Factores Inmunológicos , Células Asesinas Naturales/inmunología , Neutrófilos/metabolismo
2.
BMC Cancer ; 19(1): 687, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-31307428

RESUMEN

BACKGROUND: In a prospective study with long-term follow-up, we analyzed circulating T cell subsets in patients with metastatic colorectal cancer (mCRC) in the context of primary tumor sidedness, KRAS status, and clinical outcome. Our primary goal was to investigate whether baseline levels of circulating T cell subsets serve as a potential biomarker of clinical outcome of mCRC patients treated with an anti-VEGF-based regimen. METHODS: The study group consisted of 36 patients with colorectal adenocarcinoma who started first-line chemotherapy with bevacizumab for metastatic disease. We quantified T cell subsets including Tregs and CD8+ T cells in the peripheral blood prior to therapy initiation. Clinical outcome was evaluated as progression-free survival (PFS), overall survival (OS), and objective response rate (ORR). RESULTS: 1) mCRC patients with KRAS wt tumors had higher proportions of circulating CD8+ cytotoxic T cells among all T cells but also higher measures of T regulatory (Treg) cells such as absolute count and a higher proportion of Tregs in the CD4+ subset. 2) A low proportion of circulating Tregs among CD4+ cells, and a high CD8:Treg ratio at initiation of VEGF-targeting therapy, were associated with favorable clinical outcome. 3) In a subset of patients with primarily right-sided mCRC, superior PFS and OS were observed when the CD8:Treg ratio was high. CONCLUSIONS: The baseline level of circulating immune cells predicts clinical outcome of 1st-line treatment with the anti-VEGF angio/immunomodulatory agent bevacizumab. Circulating immune biomarkers, namely the CD8:Treg ratio, identified patients in the right-sided mCRC subgroup with favorable outcome following treatment with 1st-line anti-VEGF treatment.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Inhibidores de la Angiogénesis/uso terapéutico , Bevacizumab/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Metástasis de la Neoplasia/tratamiento farmacológico , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Reguladores/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Adenocarcinoma/sangre , Adenocarcinoma/mortalidad , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Inhibidores de la Angiogénesis/administración & dosificación , Bevacizumab/administración & dosificación , Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Femenino , Estudios de Seguimiento , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Supervivencia sin Progresión , Estudios Prospectivos , Proteínas Proto-Oncogénicas p21(ras)/análisis , Tasa de Supervivencia
3.
Int J Cancer ; 143(6): 1483-1493, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29665011

RESUMEN

Tumor-associated macrophages can promote growth of cancers. In neuroblastoma, tumor-associated macrophages have greater frequency in metastatic versus loco-regional tumors, and higher expression of genes associated with macrophages helps to predict poor prognosis in the 60% of high-risk patients who have MYCN-non-amplified disease. The contribution of cytotoxic T-lymphocytes to anti-neuroblastoma immune responses may be limited by low MHC class I expression and low exonic mutation frequency. Therefore, we modelled human neuroblastoma in T-cell deficient mice to examine whether depletion of monocytes/macrophages from the neuroblastoma microenvironment by blockade of CSF-1R can improve the response to chemotherapy. In vitro, CSF-1 was released by neuroblastoma cells, and topotecan increased this release. In vivo, neuroblastomas formed by subcutaneous co-injection of human neuroblastoma cells and human monocytes into immunodeficient NOD/SCID mice had fewer human CD14+ and CD163+ cells and mouse F4/80+ cells after CSF-1R blockade. In subcutaneous or intra-renal models in immunodeficient NSG or NOD/SCID mice, CSF-1R blockade alone did not affect tumor growth or mouse survival. However, when combined with cyclophosphamide plus topotecan, the CSF-1R inhibitor BLZ945, either without or with anti-human and anti-mouse CSF-1 mAbs, inhibited neuroblastoma growth and synergistically improved mouse survival. These findings indicate that depletion of tumor-associated macrophages from neuroblastomas can be associated with increased chemotherapeutic efficacy without requiring a contribution from T-lymphocytes, suggesting the possibility that combination of CSF-1R blockade with chemotherapy might be effective in patients who have limited anti-tumor T-cell responses.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Animales , Apoptosis , Benzotiazoles/farmacología , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Macrófagos/inmunología , Macrófagos/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Monocitos/inmunología , Monocitos/patología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Ácidos Picolínicos/farmacología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Neurooncol ; 138(1): 199-207, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29427149

RESUMEN

Single agent studies targeting the tumor microenvironment in central nervous system (CNS) tumors have largely been disappointing. Combination therapies targeting various pathways and cell types may be a more effective strategy. In this phase I study, we evaluated the combination of dasatinib, lenalidomide, and temozolomide in children with relapsed or refractory primary CNS tumors. Patients 1-21 years old with relapsed or refractory CNS tumors were eligible. Starting doses of dasatinib and lenalidomide were 65 mg/m2/dose twice daily and 55 mg/m2 once daily, respectively, while temozolomide was constant at 75 mg/m2 daily. The study followed a 3 + 3 phase I design, with a 4-week dose-limiting toxicity (DLT) evaluation period. Serial peripheral blood lymphocyte subsets were evaluated in consenting patients. Fifteen patients were enrolled and thirteen were DLT-evaluable. DLTs occurred in 5 patients, including somnolence and confusion (1 patient), hypokalemia (1 patient) and thrombocytopenia (3 patients). The maximum tolerated dose for the combination was dasatinib 65 mg/m2 twice daily, lenalidomide 40 mg/m2 daily, and temozolomide 75 mg/m2 daily, for 21 days followed by 7 days rest in repeating 28-day cycles. Transient increases in natural killer effector cells and cytotoxic T-cells were seen after 1 week of treatment. One out of six response-evaluable patients showed a partial response. The combination was feasible and relatively well tolerated in this heavily pre-treated population. The most common toxicities were hematologic. Preliminary evidence of clinical benefit was seen.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Adolescente , Antígenos CD/metabolismo , Neoplasias del Sistema Nervioso Central/patología , Niño , Preescolar , Terapia Combinada , Dasatinib/uso terapéutico , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Lactante , Lenalidomida/uso terapéutico , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Masculino , Temozolomida/uso terapéutico , Adulto Joven
5.
Exp Cell Res ; 334(1): 78-89, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25845499

RESUMEN

Cancer cells typically exhibit increased glycolysis and decreased mitochondrial oxidative phosphorylation, and they continue to exhibit some elevation in glycolysis even under aerobic conditions. However, it is unclear whether cancer cell lines employ a high level of glycolysis comparable to that of the original cancers from which they were derived, even if their culture conditions are changed to physiologically relevant oxygen concentrations. From three childhood acute lymphoblastic leukemia (ALL) patients we established three new pairs of cell lines in both atmospheric (20%) and physiologic (bone marrow level, 5%) oxygen concentrations. Cell lines established in 20% oxygen exhibited lower proliferation, survival, expression of glycolysis genes, glucose consumption, and lactate production. Interestingly, the effects of oxygen concentration used during cell line initiation were only partially reversible when established cell cultures were switched from one oxygen concentration to another for eight weeks. These observations indicate that ALL cell lines established at atmospheric oxygen concentration can exhibit relatively low levels of glycolysis and these levels are semi-permanent, suggesting that physiologic oxygen concentrations may be needed from the time of cell line initiation to preserve the high level of glycolysis commonly exhibited by leukemias in vivo.


Asunto(s)
Técnicas de Cultivo de Célula , Glucólisis , Oxígeno/metabolismo , Fenotipo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Animales , Línea Celular Tumoral , Colorimetría , Perfilación de la Expresión Génica , Glucosa/análisis , Glucosa/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Cancer Immunol Immunother ; 62(10): 1637-48, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23982484

RESUMEN

BACKGROUND: Treatment for children with high-risk neuroblastoma with anti-disialoganglioside mAb ch14.18, IL-2, and GM-CSF plus 13-cis-retinoic acid after myeloablative chemotherapy improves survival, but 40 % of patients still relapse during or after this therapy. The microenvironment of high-risk neuroblastoma tumors includes macrophages, IL-6, and TGFß1. We hypothesized that this microenvironment suppresses anti-tumor functions of natural killer (NK) cells and that lenalidomide, an immune-modulating drug, could overcome suppression. METHODS: Purified NK cells were cultured with IL-2, neuroblastoma/monocyte-conditioned culture medium (CM), IL-6, TGFß1, and lenalidomide in various combinations and then characterized using cytotoxicity (direct and antibody-dependent cell-mediated cytotoxicity), cytokine, flow cytometry, and Western blotting assays. Anti-tumor activity of NK cells with lenalidomide, ch14.18, or both was evaluated with a xenograft model of neuroblastoma. RESULTS: CM from neuroblastoma/monocyte co-cultures contains IL-6 and TGFß1 that suppress IL-2 activation of NK cell cytotoxicity and IFNγ secretion. IL-6 and TGFß1 activate the STAT3 and SMAD2/3 pathways in NK cells and suppress IL-2 induction of cytotoxicity, granzymes A and B release, perforin expression, and IFNγ secretion. Lenalidomide blocks IL-6 and TGFß1 activation of these signaling pathways and inhibits their suppression of NK cells. Neuroblastoma cells in NOD/SCID mice exhibit activated STAT3 and SMAD2/3 pathways. Their growth is most effectively inhibited by co-injected peripheral blood mononuclear cells (PBMC) containing NK cells when mice are treated with both ch14.18 and lenalidomide. CONCLUSION: Immunotherapy with anti-tumor cell antibodies may be improved by lenalidomide, which enhances activation of NK cells and inhibits their suppression by IL-6 and TGFß1.


Asunto(s)
Interleucina-6/farmacología , Células Asesinas Naturales/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/inmunología , Talidomida/análogos & derivados , Factor de Crecimiento Transformador beta1/farmacología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Medios de Cultivo Condicionados , Femenino , Humanos , Interleucina-6/inmunología , Células Asesinas Naturales/inmunología , Lenalidomida , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuroblastoma/metabolismo , Talidomida/farmacología , Factor de Crecimiento Transformador beta1/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
7.
Sci Rep ; 13(1): 19915, 2023 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-37964011

RESUMEN

C-C motif chemokine ligand 2 (CCL2) is a monocyte chemoattractant that promotes metastatic disease and portends a poor prognosis in many cancers. To determine the potential of anti-CCL2 inhibition as a therapy for recurrent metastatic disease in neuroblastoma, a mouse model of minimal residual disease was utilized in which residual disease was treated with anti-CCL2 monoclonal antibody with etoposide. The effect of anti-CCL2 antibody on neuroblastoma cells was determined in vitro with cell proliferation, transwell migration, and 2-dimensional chemotaxis migration assays. The in vivo efficacy of anti-CCL2 antibody and etoposide against neuroblastoma was assessed following resection of primary tumors formed by two cell lines or a patient-derived xenograft (PDX) in immunodeficient NOD-scid gamma mice. In vitro, anti-CCL2 antibody did not affect cell proliferation but significantly inhibited neuroblastoma cell and monocyte migration towards an increasing CCL2 concentration gradient. Treatment of mice with anti-CCL2 antibody combined with etoposide significantly increased survival of mice after resection of primary tumors, compared to untreated mice.


Asunto(s)
Neuroblastoma , Humanos , Animales , Ratones , Etopósido/farmacología , Etopósido/uso terapéutico , Ligandos , Neoplasia Residual/tratamiento farmacológico , Ratones Endogámicos NOD , Neuroblastoma/patología , Quimiocinas , Quimiocina CCL2 , Línea Celular Tumoral
8.
Oncoimmunology ; 10(1): 1838140, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33489468

RESUMEN

Immune checkpoint therapy has resulted in minimal clinical response in many pediatric cancers. We sought to understand the influence of immune checkpoint inhibition using anti-PD-1 and anti-CTLA-4 antibodies individually, in combination, and after chemotherapy on immune responses in minimal and established murine neuroblastoma models. We also sought to understand the role of the tumor microenvironment (TME) and PD-L1 expression and their alteration post-chemotherapy in our models and human tissues. PD-L1 expression was enriched in human tumor-associated macrophages and up-regulated after chemotherapy. In a murine minimal disease model, single and dual immune checkpoint blockade promoted tumor rejection, improved survival, and established immune memory with long-term anti-tumor immunity against re-challenge. In an established tumor model, only dual immune checkpoint blockade showed efficacy. Interestingly, dual immune checkpoint therapy distinctly influenced adaptive and innate immune responses, with significant increase in CD8+CD28+PD-1+ T cells and inflammatory macrophages (CD11bhiCD11c-F4/80+Ly6Chi) in tumor-draining lymph nodes. Adding chemotherapy before immunotherapy provided significant survival benefit for mice with established tumors receiving anti-PD-1 or dual immune checkpoint blockade. Our findings demonstrate anti-PD-1 and anti-CTLA-4 therapy induces a novel subset of effector T cells, and support administration of induction chemotherapy immediately prior to immune checkpoint blockade in children with high-risk neuroblastoma.


Asunto(s)
Neuroblastoma , Receptor de Muerte Celular Programada 1 , Animales , Antígenos CD28 , Linfocitos T CD8-positivos , Humanos , Inhibidores de Puntos de Control Inmunológico , Ratones , Neuroblastoma/tratamiento farmacológico , Linfocitos T , Microambiente Tumoral
9.
J Immunother Cancer ; 8(2)2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33428582

RESUMEN

BACKGROUND: Immunotherapy with anti-disialoganglioside dinutuximab has improved survival for children with high-risk neuroblastoma (NB) when given after induction chemotherapy and surgery. However, disease recurrence and resistance persist. Dinutuximab efficacy has not been evaluated when initiated before primary tumor removal. Using a surgical mouse model of human NB, we examined if initiating dinutuximab plus ex vivo-activated natural killer (aNK) cells before resection of the primary tumor improves survival. METHODS: In vitro, human NB cells (SMS-KCNR-Fluc, CHLA-255-Fluc) were treated with dinutuximab and/or aNK cells and cytotoxicity was measured. In vivo, NB cells (SMS-KCNR-Fluc, CHLA-255-Fluc, or COG-N-415x PDX) were injected into the kidney of NOD-scid gamma mice. Mice received eight intravenous infusions of aNK cells plus dinutuximab beginning either 12 days before or 2 days after resection of primary tumors. Tumors in control mice were treated by resection alone or with immunotherapy alone. Disease was quantified by bioluminescent imaging and survival was monitored. aNK cell infiltration into primary tumors was quantified by flow cytometry and immunohistochemistry at varying timepoints. RESULTS: In vitro, aNK cells and dinutuximab were more cytotoxic than either treatment alone. In vivo, treatment with aNK cells plus dinutuximab prior to resection of the primary tumor was most effective in limiting metastatic disease and prolonging survival. aNK cell infiltration into xenograft tumors was observed after 1 day and peaked at 5 days following injection. CONCLUSION: Dinutuximab plus aNK cell immunotherapy initiated before resection of primary tumors decreases disease burden and prolongs survival in an experimental mouse model of NB. These findings support the clinical investigation of this treatment strategy during induction therapy in patients with high-risk NB.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neuroblastoma/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Humanos , Ratones , Neuroblastoma/mortalidad , Análisis de Supervivencia
10.
Clin Cancer Res ; 25(15): 4761-4774, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31068371

RESUMEN

PURPOSE: We determined whether elimination of CD105+ cells in the tumor microenvironment (TME) with anti-CD105 antibodies enhanced anti-disialoganglioside (GD2) antibody dinutuximab therapy of neuroblastoma when combined with activated natural killer (aNK) cells. EXPERIMENTAL DESIGN: The effect of MSCs and monocytes on antibody-dependent cellular cytotoxicity (ADCC) mediated by dinutuximab with aNK cells against neuroblastoma cells was determined in vitro. ADCC with anti-CD105 mAb TRC105 and aNK cells against MSCs, monocytes, and endothelial cells, which express CD105, was evaluated. Anti-neuroblastoma activity in immunodeficient NSG mice of dinutuximab with aNK cells without or with anti-CD105 mAbs was determined using neuroblastoma cell lines and a patient-derived xenograft. RESULTS: ADCC mediated by dinutuximab with aNK cells against neuroblastoma cells in vitro was suppressed by addition of MSCs and monocytes, and dinutuximab with aNK cells was less effective against neuroblastomas formed with coinjected MSCs and monocytes in NSG mice than against those formed by tumor cells alone. Anti-CD105 antibody TRC105 with aNK cells mediated ADCC against MSCs, monocytes, and endothelial cells. Neuroblastomas formed in NSG mice by two neuroblastoma cell lines or a patient-derived xenograft coinjected with MSCs and monocytes were most effectively treated with dinutuximab and aNK cells when anti-human (TRC105) and anti-mouse (M1043) CD105 antibodies were added, which depleted human MSCs and murine endothelial cells and macrophages from the TME. CONCLUSIONS: Immunotherapy of neuroblastoma with anti-GD2 antibody dinutuximab and aNK cells is suppressed by CD105+ cells in the TME, but suppression is overcome by adding anti-CD105 antibodies to eliminate CD105+ cells.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antineoplásicos/farmacología , Endoglina/antagonistas & inhibidores , Gangliósidos/antagonistas & inhibidores , Inmunoterapia/métodos , Células Asesinas Naturales/inmunología , Neuroblastoma/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Línea Celular Tumoral , Endoglina/inmunología , Gangliósidos/inmunología , Humanos , Células Asesinas Naturales/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuroblastoma/inmunología , Neuroblastoma/metabolismo , Neuroblastoma/patología , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Clin Cancer Res ; 25(1): 325-333, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30232225

RESUMEN

PURPOSE: Immunotherapy of neuroblastoma that remains after myeloablative chemotherapy with anti-GD2 antibody dinutuximab has increased the two-year event-free and overall survival of high-risk neuroblastoma patients; however, 40% of patients develop recurrent disease during or after this treatment. To determine the potential of such antibody-based immunotherapy earlier in treatment, a mouse model was developed in which surgical resection of the primary tumor was followed by therapy of residual disease with dinutuximab combined with ex vivo-activated human natural killer (aNK) cells. EXPERIMENTAL DESIGN: The effect of combining dinutuximab with human aNK cells was determined in vitro with cellular cytotoxicity and Matrigel invasion assays. The in vivo efficacy of dinutuximab and aNK cells against neuroblastoma was assessed following resection of primary tumors formed by two cell lines or a patient-derived xenograft (PDX) in immunodeficient NOD-scid gamma mice. RESULTS: In vitro, the combination of aNK cells and dinutuximab caused cytotoxicity and decreased invasiveness of three human neuroblastoma cell lines. Treatment of mice with dinutuximab combined with aNK cells after surgical resection of primary intrarenal tumors formed by two cell lines or a PDX decreased tumor cells in liver and bone marrow as evaluated by histopathology and bioluminescence imaging. Survival of mice after resection of these tumors was most significantly increased by treatment with dinutuximab combined with aNK cells compared with that of untreated mice. CONCLUSIONS: The combination of dinutuximab and adoptively transferred human aNK cells following surgical resection of primary neuroblastomas significantly improves survival of immunodeficient mice.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Anticuerpos Monoclonales/farmacología , N-Acetilgalactosaminiltransferasas/genética , Neuroblastoma/terapia , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Terapia Combinada , Citotoxicidad Inmunológica/efectos de los fármacos , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Inmunoterapia , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Ratones , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , Neuroblastoma/inmunología , Neuroblastoma/cirugía
12.
Cancer Res ; 77(18): 5142-5157, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28687621

RESUMEN

Cancer-associated fibroblasts (CAF) have been suggested to originate from mesenchymal stromal cells (MSC), but their relationship with MSCs is not clear. Here, we have isolated from primary human neuroblastoma tumors a population of αFAP- and FSP-1-expressing CAFs that share phenotypic and functional characteristics with bone marrow-derived MSCs (BM-MSC). Analysis of human neuroblastoma tumors also confirmed the presence of αFAP- and FSP-1-positive cells in the tumor stroma, and their presence correlated with that of M2 tumor-associated macrophages. These cells (designated CAF-MSCs) enhanced in vitro neuroblastoma cell proliferation, survival, and resistance to chemotherapy and stimulated neuroblastoma tumor engraftment and growth in immunodeficient mice, indicating an effect independent of the immune system. The protumorigenic activity of MSCs in vitro and in xenografted mice was dependent on the coactivation of JAK2/STAT3 and MEK/ERK1/2 in neuroblastoma cells. In a mouse model of orthotopically implanted neuroblastoma cells, inhibition of JAK2/STAT3 and MEK/ERK/1/2 by ruxolitinib and trametinib potentiated tumor response to etoposide and increased overall survival. These data point to a new type of protumorigenic CAF in the tumor microenvironment of neuroblastoma and to STAT3 and ERK1/2 as mediators of their activity. Cancer Res; 77(18); 5142-57. ©2017 AACR.


Asunto(s)
Antineoplásicos/farmacología , Fibroblastos Asociados al Cáncer/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Madre Mesenquimatosas/patología , Neuroblastoma/patología , Animales , Apoptosis/efectos de los fármacos , Biomarcadores de Tumor/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Janus Quinasa 2/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Masculino , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Nitrilos , Pirazoles/farmacología , Piridonas/farmacología , Pirimidinas , Pirimidinonas/farmacología , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Cancer Res ; 23(3): 804-813, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27756784

RESUMEN

PURPOSE: Immunotherapy of high-risk neuroblastoma using the anti-GD2 antibody dinutuximab induces antibody-dependent cell-mediated cytotoxicity (ADCC). Galunisertib, an inhibitor of TGFßR1, was examined for its ability to enhance the efficacy of dinutuximab in combination with human ex vivo activated NK (aNK) cells against neuroblastoma. EXPERIMENTAL DESIGN: TGFB1 and TGFBR1 mRNA expression was determined for 249 primary neuroblastoma tumors by microarray analysis. The ability of galunisertib to inhibit SMAD activity induced by neuroblastoma patient blood and bone marrow plasmas in neuroblastoma cells was tested. The impact of galunisertib on TGFß1-induced inhibition of aNK cytotoxicity and ADCC in vitro and on anti-neuroblastoma activity in NOD-scid gamma (NSG) mice was determined. RESULTS: Neuroblastomas express TGFB1 and TGFBR1 mRNA. Galunisertib suppressed SMAD activation in neuroblastoma cells induced by exogenous TGFß1 or by patient blood and bone marrow plasma, and suppressed SMAD2 phosphorylation in human neuroblastoma cells growing in NSG mice. In NK cells treated in vitro with exogenous TGFß1, galunisertib suppressed SMAD2 phosphorylation and restored the expression of DNAM-1, NKp30, and NKG2D cytotoxicity receptors and the TRAIL death ligand, the release of perforin and granzyme A, and the direct cytotoxicity and ADCC of aNK cells against neuroblastoma cells. Addition of galunisertib to adoptive cell therapy with aNK cells plus dinutuximab reduced tumor growth and increased survival of mice injected with two neuroblastoma cell lines or a patient-derived xenograft. CONCLUSIONS: Galunisertib suppresses activation of SMAD2 in neuroblastomas and aNK cells, restores NK cytotoxic mechanisms, and increases the efficacy of dinutuximab with aNK cells against neuroblastoma tumors. Clin Cancer Res; 23(3); 804-13. ©2016 AACRSee related commentary by Zenarruzabeitia et al., p. 615.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Células Asesinas Naturales/trasplante , Proteínas de Neoplasias/antagonistas & inhibidores , Neuroblastoma/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirazoles/farmacología , Quinolinas/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/fisiología , Animales , Antineoplásicos Inmunológicos/farmacología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunoterapia Adoptiva , Masculino , Ratones , Ratones Endogámicos NOD , Proteínas de Neoplasias/fisiología , Neuroblastoma/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/biosíntesis , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteína Smad2/antagonistas & inhibidores , Proteína Smad2/metabolismo , Organismos Libres de Patógenos Específicos , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Oncotarget ; 8(53): 91516-91529, 2017 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-29207662

RESUMEN

Tumor-associated macrophages (TAMs) are strongly associated with poor survival in neuroblastomas that lack MYCN amplification. To study TAM action in neuroblastomas, we used a novel murine model of spontaneous neuroblastoma lacking MYCN amplification, and observed recruitment and polarization of TAMs, which in turn enhanced neuroblastoma proliferation and growth. In both murine and human neuroblastoma cells, we found that TAMs increased STAT3 activation in neuroblastoma cells and transcriptionally up-regulated the MYC oncogene. Analysis of human neuroblastoma tumor specimens revealed that MYC up-regulation correlates with markers of TAM infiltration. In an IL6ko neuroblastoma model, the absence of IL-6 protein had no effect on tumor development and prevented neither STAT3 activation nor MYC up-regulation. In contrast, inhibition of JAK-STAT activation using AZD1480 or the clinically admissible inhibitor ruxolitinib significantly reduced TAM-mediated growth of neuroblastomas implanted subcutaneously in NOD scid gamma mice. Our results point to a unique mechanism in which TAMs promote tumor cells that lack amplification of an oncogene common to the malignancy by up-regulating transcriptional expression of a distinct oncogene from the same gene family, and underscore the role of IL-6-independent activation of STAT3 in this mechanism. Amplification of MYCN or constitutive up-regulation of MYC protein is observed in approximately half of high-risk tumors; our findings indicate a novel role of TAMs as inducers of MYC expression in neuroblastomas lacking independent oncogene activation.

15.
Leuk Res ; 30(1): 81-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15998540

RESUMEN

Dimethyl sulfoxide (DMSO) is a widely used prototypical chemical inducer of cell differentiation. In the present study, the effects of DMSO on susceptibility of human myeloid leukemia U937 cells towards ligation of distinct death receptors (DRs) were investigated. DMSO sensitized cells towards induction of apoptosis by anti-Fas antibody, tumour necrosis factor-alpha or Apo2 ligand/TNF-related apoptosis-inducing ligand (TRAIL). Apart from increasing Fas levels, DMSO did not affect expression of proteins in death signal transduction, such as Bcl-2 family proteins, FADD, caspase-3 and -8, the inhibitor of apoptosis proteins (IAPs) or cFLIP(L). However, DMSO significantly potentiated mitochondrial membrane depolarization, suggesting that this mechanism might be involved in sensitisation of myeloid cells to DR-mediated apoptosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Crioprotectores/farmacología , Dimetilsulfóxido/farmacología , Leucemia Mieloide/metabolismo , Glicoproteínas de Membrana/metabolismo , Membranas Mitocondriales/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Caspasa 3 , Caspasa 8 , Caspasas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Leucemia Mieloide/patología , Mitocondrias/metabolismo , Mitocondrias/patología , Membranas Mitocondriales/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Células U937
16.
Cancer Res ; 63(21): 7176-84, 2003 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-14612511

RESUMEN

The p53 tumor suppressor protein is known to regulate the expression of the CD95 (Fas/APO-1) death receptor in a small subset of normal cell types as well as in many cancer cell types. However, whether p53-dependent regulation of CD95 expression is consistently associated with increased susceptibility to CD95-mediated cell death is poorly understood. To address this issue, we examined constitutive and induced CD95 surface expression and function in wild-type p53-expressing carcinoma cells relative to their isogenic p53-inactivated counterparts. We compared HCT116 colorectal carcinoma cells with their p53 biallelic knock-outs and control-transfected MCF-7 breast carcinoma cells with MCF-7 cells expressing a miniprotein inhibitor of p53 (p53DD). In both cell lines, the constitutive expression of surface CD95 was significantly reduced in p53-inactivated cells, as was the apoptotic response to agonistic anti-CD95 antibody. In both cell lines, only cells with wild-type p53 activity exhibited up-regulation of surface CD95 after ionizing irradiation. Interestingly, induction of CD95 expression substantially enhanced the apoptotic response to CD95 ligation only in MCF-7 cells but not in HCT116 cells. These findings provide direct evidence for a major role for wild-type p53 activity in regulating constitutive expression and function of CD95 in carcinoma cells; however, they also demonstrate that the functional effect of DNA damage-induced up-regulation of CD95 may be cell type specific.


Asunto(s)
Proteína p53 Supresora de Tumor/fisiología , Receptor fas/fisiología , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Daño del ADN/fisiología , Silenciador del Gen , Células HCT116 , Humanos , Transfección , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/fisiología , Regulación hacia Arriba/efectos de la radiación , Rayos X , Receptor fas/biosíntesis , Receptor fas/genética
17.
Oncogene ; 21(2): 190-7, 2002 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-11803462

RESUMEN

Many viruses are known to disarm or suppress the cell death machinery of infected cells. Apoptotic cell death can be activated by aggregation of the CD95 cell surface death receptor in sensitive cells, and in most insensitive cells treated with sensitizing agents such as interferon-gamma or inhibitors of protein synthesis. We show that, subsequent to sequestration and inactivation of the p53 tumour suppressor protein, SV40 abrogates p53-dependent, DNA damage-inducible up-regulation of CD95 surface expression. Loss of surface up-regulation of CD95 after sub-lethal mitomycin C treatment resulted in an impaired enhancement of both caspase-8 cleavage and apoptotic cell death following CD95 aggregation. We conclude that infection of human cells with SV40 virus strongly inhibits DNA damage-induced enhancement of CD95-mediated apoptosis.


Asunto(s)
Daño del ADN , Virus 40 de los Simios/fisiología , Receptor fas/genética , Adenocarcinoma , Anticuerpos Monoclonales , Apoptosis , Neoplasias de la Mama , Ciclo Celular/genética , Femenino , Humanos , Inmunoglobulina G , Células Tumorales Cultivadas , Receptor fas/inmunología
18.
Clin Cancer Res ; 19(8): 2132-43, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23378384

RESUMEN

PURPOSE: Adoptive transfer of natural killer (NK) cells combined with tumor-specific monoclonal antibodies (mAb) has therapeutic potential for malignancies. We determined if large numbers of activated NK (aNK) cells can be grown ex vivo from peripheral blood mononuclear cells (PBMC) of children with high-risk neuroblastoma using artificial antigen-presenting cells (aAPC). EXPERIMENTAL DESIGN: Irradiated K562-derived Clone 9.mbIL21 aAPC were cocultured with PBMC, and propagated NK cells were characterized with flow cytometry, cytotoxicity assays, Luminex multicytokine assays, and a nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse model of disseminated neuroblastoma. RESULTS: Coculturing patient PBMC with aAPC for 14 days induced 2,363- ± 443-fold expansion of CD56(+)CD3(-)CD14(-) NK cells with 83% ± 3% purity (n = 10). Results were similar to PBMC from normal donors (n = 5). Expression of DNAM-1, NKG2D, FcγRIII/CD16, and CD56 increased 6- ± 3-, 10- ± 2-, 21- ± 20-, and 18- ± 3-fold, respectively, on day 14 compared with day 0, showing activation of NK cells. In vitro, aNK cells were highly cytotoxic against neuroblastoma cell lines and killing was enhanced with GD2-specific mAb ch14.18. When mediating cytotoxicity with ch14.18, release of TNF-α, granulocyte macrophage colony-stimulating factor, IFN-γ, sCD40L, CCL2/MCP-1, CXCL9/MIG, and CXCL11/I-TAC by aNK cells increased 4-, 5-, 6-, 15-, 265-, 917-, and 363-fold (151-9,121 pg/mL), respectively, compared with aNK cells alone. Survival of NOD/SCID mice bearing disseminated neuroblastoma improved when treated with thawed and immediately intravenously infused cryopreserved aNK cells compared with untreated mice and was further improved when ch14.18 was added. CONCLUSION: Propagation of large numbers of aNK cells that maintain potent antineuroblastoma activities when cryopreserved supports clinical testing of adoptive cell therapy with ch14.18.


Asunto(s)
Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/inmunología , Neuroblastoma/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Antígeno CD56/inmunología , Antígeno CD56/metabolismo , Línea Celular Tumoral , Células Cultivadas , Niño , Técnicas de Cocultivo , Citocinas/inmunología , Citocinas/metabolismo , Citotoxicidad Inmunológica/inmunología , Citometría de Flujo , Humanos , Inmunoterapia Adoptiva/métodos , Células K562 , Estimación de Kaplan-Meier , Células Asesinas Naturales/metabolismo , Células Asesinas Naturales/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neuroblastoma/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Immunother ; 36(5): 319-29, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23719242

RESUMEN

Neuroblastoma cells have been reported to be resistant to death induced by soluble, recombinant forms of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) (CD253/TNFSF10) because of low or absent expression of caspase-8 and/or TRAIL-receptor 2 (TRAIL-R2/DR5/CD262/TNFRSF10b). However, their sensitivity to membrane-bound TRAIL on natural killer (NK) cells is not known. Comparing microarray gene expression and response to NK cell-mediated cytotoxicity, we observed a correlation between TRAIL-R2 expression and the sensitivity of 14 neuroblastoma cell lines to the cytotoxicity of NK cells activated with interleukin (IL)-2 plus IL-15. Even though most NK cytotoxicity was dependent upon perforin, the cytotoxicity was supplemented by TRAIL in 14 of 17 (82%) neuroblastoma cell lines as demonstrated using an anti-TRAIL neutralizing antibody. Similarly, a recently developed NK cell expansion system employing IL-2 plus lethally irradiated K562 feeder cells constitutively expressing membrane-bound IL-21 (K562 clone 9.mbIL21) resulted in activated NK cells derived from normal healthy donors and neuroblastoma patients that also utilized TRAIL to supplement cytotoxicity. Exogenous interferon-γ upregulated expression of caspase-8 in 3 of 4 neuroblastoma cell lines and increased the contribution of TRAIL to NK cytotoxicity against 2 of the 3 lines; however, relatively little inhibition of cytotoxicity was observed when activated NK cells were treated with an anti-interferon-γ neutralizing antibody. Constraining the binding of anti-TRAIL neutralizing antibody to membrane-bound TRAIL but not soluble TRAIL indicated that membrane-bound TRAIL alone was responsible for essentially all of the supplemental cytotoxicity. Together, these findings support a role for membrane-bound TRAIL in the cytotoxicity of NK cells against neuroblastoma cells.


Asunto(s)
Células Asesinas Naturales/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Neuroblastoma/tratamiento farmacológico , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Anticuerpos Bloqueadores/farmacología , Apoptosis/efectos de los fármacos , Caspasa 8/genética , Caspasa 8/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Citocinas/inmunología , Citotoxicidad Inmunológica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células K562 , Células Asesinas Naturales/inmunología , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Membrana/inmunología , Análisis por Micromatrices , Neuroblastoma/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Transcriptoma
20.
Cancer Res ; 73(13): 3852-64, 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633489

RESUMEN

Drug resistance is a major cause of treatment failure in cancer. Here, we have evaluated the role of STAT3 in environment-mediated drug resistance (EMDR) in human neuroblastoma. We determined that STAT3 was not constitutively active in most neuroblastoma cell lines but was rapidly activated upon treatment with interleukin (IL)-6 alone and in combination with the soluble IL-6 receptor (sIL-6R). Treatment of neuroblastoma cells with IL-6 protected them from drug-induced apoptosis in a STAT3-dependent manner because the protective effect of IL-6 was abrogated in the presence of a STAT3 inhibitor and upon STAT3 knockdown. STAT3 was necessary for the upregulation of several survival factors such as survivin (BIRC5) and Bcl-xL (BCL2L1) when cells were exposed to IL-6. Importantly, IL-6-mediated STAT3 activation was enhanced by sIL-6R produced by human monocytes, pointing to an important function of monocytes in promoting IL-6-mediated EMDR. Our data also point to the presence of reciprocal activation of STAT3 between tumor cells and bone marrow stromal cells including not only monocytes but also regulatory T cells (Treg) and nonmyeloid stromal cells. Thus, the data identify an IL-6/sIL-6R/STAT3 interactive pathway between neuroblastoma cells and their microenvironment that contributes to drug resistance.


Asunto(s)
Neoplasias Óseas/metabolismo , Resistencia a Antineoplásicos , Interleucina-6/fisiología , Neuroblastoma/metabolismo , Factor de Transcripción STAT3/fisiología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Etopósido/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Melfalán/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Monocitos/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Receptores de Interleucina-6/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA