Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Br J Cancer ; 130(4): 671-681, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38148376

RESUMO

BACKGROUND: Recent studies have identified that low levels of some tumour suppressor microRNAs (miRNAs) in the blood contribute to tumour progression and poor outcomes in various cancers. However, no study has proved these miRNAs are associated with cancer immune mechanisms. METHODS: From a systematic review of the NCBI and miRNA databases, four tumour suppressor miRNA candidates were selected (miR-5193, miR-4443, miR-520h, miR-496) that putatively target programmed cell death ligand 1 (PD-L1). RESULTS: Test-scale and large-scale analyses revealed that plasma levels of miR-5193 were significantly lower in gastric cancer (GC) patients than in healthy volunteers (HVs). Low plasma levels of miR-5193 were associated with advanced pathological stages and were an independent prognostic factor. Overexpression of miR-5193 in GC cells suppressed PD-L1 on the surface of GC cells, even with IFN-γ stimulation. In the coculture model of GC cells and T cells stimulated by anti-CD3/anti-CD28 beads, overexpression of miR-5193 increased anti-tumour activity of T cells by suppressing PD-L1 expression. Subcutaneous injection of miR-5193 also significantly enhanced the tumour-killing activity and trafficking of T cells in mice. CONCLUSIONS: Low blood levels of miR-5193 are associated with GC progression and poor outcomes and could be a target of nucleic acid immunotherapy in GC patients.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Animais , Camundongos , Neoplasias Gástricas/genética , Neoplasias Gástricas/terapia , Antígeno B7-H1 , MicroRNAs/metabolismo , Genes Supressores de Tumor , Imunoterapia
2.
Cancer Immunol Immunother ; 72(4): 957-968, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36214866

RESUMO

CD19-specific chimeric antigen receptor T (CAR T) immunotherapy is used to treat B-cell malignancies. However, antigen-escape mediated relapse following CAR T therapy has emerged as a major concern. In some relapsed cases, especially KMT2A rearrangement-positive B-acute lymphoblastic leukemia (KMT2A-r B-ALL), most of the B-cell antigens are lost via lineage conversion to the myeloid phenotype, rendering multi-B-cell-antigen-targeted CAR T cell therapy ineffective. Fms-related tyrosine kinase-3 (FLT3) is highly expressed in KMT2A-r B-ALL; therefore, in this study, we aimed to evaluate the antitumor efficacy of CAR T cells targeting both CD19 and FLT3 in KMT2A-r B-ALL cells. We developed piggyBac transposon-mediated CAR T cells targeting CD19, FLT3, or both (dual) and generated CD19-negative KMT2A-r B-ALL models through CRISPR-induced CD19 gene-knockout (KO). FLT3 CAR T cells showed antitumor efficacy against CD19-KO KMT2A-r B-ALL cells both in vitro and in vivo; dual-targeted CAR T cells showed cytotoxicity against wild-type (WT) and CD19-KO KMT2A-r B-ALL cells, whereas CD19 CAR T cells demonstrated cytotoxicity only against WT KMT2A-r B-ALL cells in vitro. Therefore, targeting FLT3-specific CAR T cells would be a promising strategy for KMT2A-r B-ALL cells even with CD19-negative relapsed cases.


Assuntos
Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Humanos , Antígenos CD19/genética , Tirosina Quinase 3 Semelhante a fms/genética , Imunoterapia , Imunoterapia Adotiva , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos Quiméricos/genética , Linfócitos T , Proteína de Leucina Linfoide-Mieloide/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo
3.
Int J Clin Oncol ; 28(6): 736-747, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36859566

RESUMO

Since the introduction of the use of chimeric antigen receptor T-cell therapy (CAR-T therapy) dramatically changed the therapeutic strategy for B cell tumors, various CAR-T cell products have been developed and applied to myeloid and solid tumors. Although viral vectors have been widely used to produce genetically engineered T cells, advances in genetic engineering have led to the development of methods for producing non-viral, gene-modified CAR-T cells. Recent progress has revealed that non-viral CAR-T cells have a significant impact not only on the simplicity of the production process and the accessibility of non-viral vectors but also on the function of the cells themselves. In this review, we focus on piggyBac-transposon-based CAR-T cells among non-viral, gene-modified CAR-T cells and discuss their characteristics, preclinical development, and recent clinical applications.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T , Humanos , Receptores de Antígenos de Linfócitos T/genética , Neoplasias/patologia , Linfócitos T , Imunoterapia Adotiva/métodos
4.
Rinsho Ketsueki ; 64(5): 418-426, 2023.
Artigo em Japonês | MEDLINE | ID: mdl-37271534

RESUMO

The clinical application of chimeric antigen receptor T-cell therapy (CAR-T therapy) has significantly altered the therapeutic strategy for B-cell tumors and is now being used to treat myeloid and solid tumors. Nonetheless, the efficacy of CAR-T cell therapy for myeloid and solid tumors has been limited, and several studies are being conducted to understand and overcome the underlying mechanisms. Recent research achievements have revealed that the properties of CAR-T cells, particularly their memory function, which can be continuously amplified in the body without exhaustion after administration, are closely related to CAR-T cell clinical efficacy. Furthermore, because the characteristics of CAR-T cells are greatly influenced by the quality of peripheral blood mononuclear cells, the raw material of CAR-T cells, and the T-cell used during the manufacturing process, attention has been drawn to the development of high-quality CAR-T cell manufacturing methods and combination therapies that maintain CAR-T cell memory function and suppress immune exhaustion. This article provides an overview of the current state of CAR-T cell development and clinical application to cancer, particularly emphasizing the development of manufacturing processes and efforts to improve CAR-T cell efficacy in combination therapy with molecular-targeting drugs.


Assuntos
Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Leucócitos Mononucleares/patologia , Imunoterapia Adotiva/métodos , Linfócitos T , Neoplasias/genética , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética
5.
Biochem Biophys Res Commun ; 635: 144-153, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36274364

RESUMO

Breast cancer is a complex heterogeneous disease with unique molecular subtypes, which limits the development of optimized treatment strategies for each subtype. Cancer gene therapy and potential therapeutics for advanced/refractory cancers can be promising for breast cancer. Combining tumor-tropic lipid nanoparticles (LNPs) and inducible caspase-9 (iC9) mRNA, we aimed to develop a novel treatment strategy for refractory breast cancer. LNP's anti-tumor effects were tested in vitro in three breast cancer cell lines: MDA-MB231, SKBR3, and MCF-7. Tumor cells were treated with LNPs encapsulated with eGFP or iC9 mRNA and chemical inducers of dimerization (CID). Apoptosis-related genes were evaluated by reverse transcriptase quantitative PCR. LNPs could efficiently deliver encapsulated GFP mRNA to all three cancer cell lines (>80% GFP expression. in target cells). Furthermore, LNPs encapsulated with iC9 mRNA (iC9-LNPs) and CID showed cytotoxic activity against all cancer cell lines in vitro. Interestingly, susceptibility to iC9 gene therapy was heterogeneous among cancer cell lines. iC9-LNPs with CID-induced potent cytotoxic effects against SKBR3 and MDA-MB231 cells, but only a mild cytotoxic effect on MCF7 cells. Quantification of apoptosis-related genes suggested that a high BAX/Bcl-2 ratio might be associated with iC9-LNP + CID susceptibility. Thus, cancer gene therapy using iC9-LNPs and CID could be a promising alternative for the treatment of breast cancers, especially for aggressive breast cancers.


Assuntos
Antineoplásicos , Neoplasias da Mama , Nanopartículas , Humanos , Feminino , Caspase 9/genética , Caspase 9/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , RNA Mensageiro/genética , Nanopartículas/química
6.
Pediatr Int ; 64(1): e15159, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35727889

RESUMO

BACKGROUND: Patients with high-risk neuroblastoma have a poor prognosis; new therapeutic agents are therefore required. We investigated the antitumor effects of OBP-801, a novel histone deacetylase inhibitor, its underlying mechanism, and its potential as a therapeutic agent for patients with neuroblastoma. METHODS: The study included five human neuroblastoma cell lines: IMR32, GOTO, KP-N-RTBM, SK-N-AS, and SH-SY5Y. We investigated cell proliferation, cell cycle status, protein expression patterns, and apoptosis in neuroblastoma cells after OBP-801 treatment in vitro. Cell survival rate and cell cycle were analyzed using the WST-8 assay and flow cytometry, respectively. Apoptosis was detected using annexin V staining, and the expression of apoptosis-related proteins was investigated by western blotting. The antitumor activity of OBP-801 was examined in an in vivo xenograft mouse model. RESULTS: Dose-effect curve analysis showed that the mean half-maximal inhibitory concentration value was 5.5 ± 5.9 nM for the MYCN-amplified cell lines (IMR32, GOTO, and KP-N-RTBM) and 3.1 ± 0.7 nM for the MYCN-nonamplified cell lines (SK-N-AS and SH-SY5Y). OBP-801 inhibited cell proliferation and growth in all the cell lines. It induced G2/M phase arrest through the p21 (CDKN1A) pathway, increasing histone H3 levels and, subsequently, apoptosis in human neuroblastoma cells. OBP-801 suppressed the growth of neuroblastoma cells in the mouse xenograft model. CONCLUSIONS: Overall, OBP-801 induces M-phase arrest and apoptosis in neuroblastoma cells via mitotic catastrophe. Our results indicate that OBP-801 is a promising therapeutic agent with fewer adverse effects for patients with neuroblastoma.


Assuntos
Neuroblastoma , Animais , Linhagem Celular Tumoral , Proliferação de Células , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Camundongos , Proteína Proto-Oncogênica N-Myc/uso terapêutico , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico
7.
Pediatr Surg Int ; 39(1): 43, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36484857

RESUMO

PURPOSE: Mouse IgG anti-disialoganglioside GD2 antibody-secreting mouse mesenchymal stem cells (anti-GD2-MSCs) were developed, and their anti-tumor effects were validated in an in vivo neuroblastoma mouse model. METHODS: Anti-GD2 antibody constructs were generated, incorporating FLAG-tagged single-chain fragment variables against GD2 fused to a linker sequence, and a fragment of a stationary portion was changed from human IgG to mouse IgG and GFP protein. The construct was lentivirally introduced into mouse MSCs. A syngeneic mouse model was established through the subcutaneous transplantation of a tumor tissue fragment from a TH-MYCN transgenic mouse, and the homing effects of anti-GD2-MSCs were validated by In vivo imaging system imaging. The syngeneic model was divided into three groups according to topical injection materials: anti-GD2-MSCs with IL-2, IL-2, and PBS. The tumors were removed, and natural killer (NK) cells were counted. RESULTS: Anti-GD2-MSCs showed homing effects in syngeneic models. The growth rate of subcutaneous tumors was significantly suppressed by anti-GD2-MSCs with IL-2 (p < 0.05). Subcutaneous tumor immunostaining showed an increased NK cell infiltration in the same group (p < 0.01). CONCLUSION: Anti-GD2-MSCs using mouse IgG showed a homing effect and significant tumor growth suppression in syngeneic models. Anti-GD2-MSC-based cellular immunotherapy could be a novel therapeutic strategy for intractable neuroblastoma.


Assuntos
Células-Tronco Mesenquimais , Neuroblastoma , Humanos , Camundongos , Animais , Gangliosídeos/uso terapêutico , Interleucina-2/uso terapêutico , Neuroblastoma/metabolismo , Modelos Animais de Doenças , Camundongos Transgênicos , Imunoglobulina G/uso terapêutico
8.
Cancer Sci ; 112(10): 4026-4036, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34382720

RESUMO

Disialoganglioside (GD2)-specific chimeric antigen receptor (CAR)-T cells (GD2-CAR-T cells) have been developed and tested in early clinical trials in patients with relapsed/refractory neuroblastoma. However, the effectiveness of immunotherapy using these cells is limited, and requires improvement. Combined therapy with CAR-T cells and molecular targeted drugs could be a promising strategy to enhance the antitumor efficacy of CAR T cell immunotherapy. Here, we generated GD2-CAR-T cells through piggyBac transposon (PB)-based gene transfer (PB-GD2-CAR-T cells), and analyzed the combined effect of these cells and a MEK inhibitor in vitro and in vivo on neuroblastoma. Trametinib, a MEK inhibitor, ameliorated the killing efficacy of PB-GD2-CAR-T cells in vitro, whereas a combined treatment of the two showed superior antitumor efficacy in a murine xenograft model compared to that of PB-GD2-CAR-T cell monotherapy, regardless of the mutation status of the MAPK pathway in tumor cells. The results presented here provide new insights into the feasibility of combined treatment with CAR-T cells and MEK inhibitors in patients with neuroblastoma.


Assuntos
Antineoplásicos/uso terapêutico , Gangliosídeos/uso terapêutico , Imunoterapia Adotiva/métodos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Neuroblastoma/terapia , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Receptores de Antígenos Quiméricos/uso terapêutico , Animais , Linhagem Celular Tumoral , Terapia Combinada/métodos , Cumarínicos/uso terapêutico , Elementos de DNA Transponíveis , Resistencia a Medicamentos Antineoplásicos , Feminino , Terapia Genética/métodos , Humanos , Camundongos , Camundongos SCID , Mutação , Recidiva Local de Neoplasia/terapia , Inibidores de Proteínas Quinases/uso terapêutico , Linfócitos T , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/antagonistas & inibidores
9.
Cancer Sci ; 112(11): 4617-4626, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34464480

RESUMO

Neuroblastoma, the most common extracranial solid tumor of childhood, is thought to arise from neural crest-derived immature cells. The prognosis of patients with high-risk or recurrent/refractory neuroblastoma remains quite poor despite intensive multimodality therapy; therefore, novel therapeutic interventions are required. We examined the expression of a cell adhesion molecule CD146 (melanoma cell adhesion molecule [MCAM]) by neuroblastoma cell lines and in clinical samples and investigated the anti-tumor effects of CD146-targeting treatment for neuroblastoma cells both in vitro and in vivo. CD146 is expressed by 4 cell lines and by most of primary tumors at any stage. Short hairpin RNA-mediated knockdown of CD146, or treatment with an anti-CD146 polyclonal antibody, effectively inhibited growth of neuroblastoma cells both in vitro and in vivo, principally due to increased apoptosis via the focal adhesion kinase and/or nuclear factor-kappa B signaling pathway. Furthermore, the anti-CD146 polyclonal antibody markedly inhibited tumor growth in immunodeficient mice inoculated with primary neuroblastoma cells. In conclusion, CD146 represents a promising therapeutic target for neuroblastoma.


Assuntos
Anticorpos/uso terapêutico , Antígeno CD146/antagonistas & inibidores , Terapia de Alvo Molecular/métodos , Neuroblastoma/terapia , RNA Interferente Pequeno/uso terapêutico , Animais , Apoptose , Antígeno CD146/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Quinase 1 de Adesão Focal/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Camundongos , NF-kappa B/metabolismo , Recidiva Local de Neoplasia , Transplante de Neoplasias , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Prognóstico , Transdução de Sinais , Esferoides Celulares , Transdução Genética/métodos
10.
Cancer Cell Int ; 20: 192, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32489328

RESUMO

BACKGROUND: Rhabdomyosarcoma (RMS) is the most common pediatric soft tissue sarcoma. There are two subtypes, fusion gene-positive RMS (FP-RMS) and fusion gene-negative RMS (FN-RMS), depending on the presence of a fusion gene, either PAX3-FOXO1 or PAX7-FOXO1. These fusion genes are thought to be oncogenic drivers of FP-RMS. By contrast, the underlying mechanism of FN-RMS has not been thoroughly investigated. It has recently been shown that HMGA2 is specifically positive in pathological tissue from FN-RMS, but the role of HMGA2 in FN-RMS remains to be clarified. METHODS: In this study, we used FN-RMS cell lines to investigate the function of HMGA2. Gene expression, cell growth, cell cycle, myogenic differentiation, tumor formation in vivo, and cell viability under drug treatment were assessed. RESULTS: We found that HMGA2 was highly expressed in FN-RMS cells compared with FP-RMS cells and that knockdown of HMGA2 in FN-RMS cells inhibited cell growth and induced G1 phase accumulation in the cell cycle and myogenic differentiation. Additionally, we showed using both gain-of-function and loss-of-function assays that HMGA2 was required for tumor formation in vivo. Consistent with these findings, the HMGA2 inhibitor netropsin inhibited the cell growth of FN-RMS. CONCLUSIONS: Our results suggest that HMGA2 has important role in the oncogenicity of FP-RMS and may be a potential therapeutic target in patients with FN-RMS.

11.
BMC Pediatr ; 20(1): 212, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398048

RESUMO

BACKGROUND: Few clinical trials have been reported for patients with intermediate-risk neuroblastoma because of the scarcity of the disease and the variety of clinical and biological characteristics. A multidisciplinary treatment that consists of multidrug chemotherapy and surgery is expected to lead to a good prognosis with few complications. Therefore, a clinical trial for patients with intermediate-risk tumors was designed to establish a standard treatment that reduces complications and achieves good outcomes. METHODS: We planned a prospective phase 2, single-arm study of the efficacy of image-defined risk factors (IDRF)-based surgical decision and stepwise treatment intensification for patients with intermediate-risk neuroblastomas. For the localized tumor group, IDRF evaluations will be performed after each three-course chemotherapy, and surgery will be performed when appropriate. For patients with metastatic tumors, a total of five chemotherapy courses will be performed, and primary lesions will be removed when the IDRF becomes negative. The primary endpoint is 3-year progression-free survival rate, and the secondary endpoints include 3-year progression-free survival rates and overall survival rates of the localized group and the metastasis group and the incidence of adverse events. From international results, 75% is considered an appropriate 3-year progression-free survival rate. If this trial's expected 3-year progression-free survival rate of 85% is statistically greater than 75% in the lower limit of the 95.3% confidence interval, with an accuracy 10% (85 ± 10%), both groups require more than 65 patients. DISCUSSION: This study is the first clinical trial on the efficacy of IDRF-based surgical decision and stepwise treatment intensification for patients with intermediate-risk neuroblastomas. We expect that this study will contribute to the establishment of a standard treatment for patients with intermediate-risk neuroblastoma. TRIAL REGISTRATION: UMIN000004700, jRCTs051180203; Registered on December 9, 2010.


Assuntos
Neuroblastoma , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Ensaios Clínicos Fase II como Assunto , Humanos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/cirurgia , Estudos Prospectivos , Fatores de Risco , Taxa de Sobrevida
12.
Biochem Biophys Res Commun ; 512(3): 524-530, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-30904164

RESUMO

Precision medicine strategies for treating rhabdomyosarcoma (RMS), a childhood malignancy, have not been developed. We examined the effect of CH5126766, a potent selective dual RAF/MEK inhibitor, on RMS cell lines. Among the eleven cell lines studied, one NRAS and two HRAS mutated cell lines were detected. CH5126766 inhibited the proliferation and growth in all of the RAS-mutated RMS cell lines, while it induced G1 cell cycle arrest in two of them. G1 cell cycle arrest was accompanied by p21 up-regulation and RB dephosphorylation. CH5126766 also suppressed the in vivo growth of RAS-mutated RMS tumor, and the mice showed improved survival. Thus, our results demonstrate that CH5126766 is an effective RAF/MEK inhibitor in RAS-mutated RMS. This study not only shows that in RMS, mutations in the RAS pathway can be a target for precision medicine, but also demonstrates that the evaluation of the gene mutation status is important in childhood malignancies.


Assuntos
Cumarínicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Rabdomiossarcoma/tratamento farmacológico , Proteínas ras/genética , Animais , Ciclo Celular , Linhagem Celular Tumoral , Criança , Cumarínicos/uso terapêutico , Feminino , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação/efeitos dos fármacos , Medicina de Precisão , Inibidores de Proteínas Quinases/uso terapêutico , Rabdomiossarcoma/genética , Transdução de Sinais/efeitos dos fármacos
13.
Pediatr Blood Cancer ; 66(11): e27914, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31342649

RESUMO

BACKGROUND: The present study sought to reduce the incidence of treatment complications of low-risk neuroblastoma by using image-defined risk factors (IDRFs) to inform the timing of surgical resection. PROCEDURES: Eligible patients included children (<18 years of age) with stage 1 or 2 disease, children (<365 days of age) with stage 3 disease, and infants with stage 4S disease. In IDRF-negative cases, treatment was completed with surgical resection alone. In IDRF-positive cases, the timing of surgery was determined based on the IDRFs after low-dose chemotherapy with 2-3 of the following four drugs: vincristine, cyclophosphamide, pirarubicin, and carboplatin. The outcome measures were overall survival, progression-free survival, and adverse events. This study was registered with the UMIN Clinical Trials Registry (number 000004355). RESULTS: Of the 60 patients screened between 2010 and 2013, 58 eligible patients were enrolled; 32 were identified as IDRF negative at diagnosis while 26 were identified as IDRF positive and underwent induction chemotherapy. The 3-year overall and progression-free survival rates of the 58 patients were 100% and 82.8% (95% confidence interval: 70.3-90.3), respectively. Neutropenia was the most frequently reported grade 3 or 4 chemotherapy-related form of toxicity (41.7%). With regard to surgical complications, 2.5% of all patients developed pleural effusion and ascites as early complications, while only 2.5% developed renal atrophy as a long-term complication. No fatal toxicities were observed. CONCLUSION: Using IDRFs to inform surgical decision making for the treatment of low-risk neuroblastoma improved prognosis and reduced the incidence of long-term complications.


Assuntos
Neuroblastoma/cirurgia , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Neutropenia Febril Induzida por Quimioterapia/etiologia , Criança , Pré-Escolar , Tomada de Decisão Clínica , Terapia Combinada , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Doxorrubicina/administração & dosagem , Doxorrubicina/efeitos adversos , Doxorrubicina/análogos & derivados , Feminino , Ganglioneuroblastoma/diagnóstico por imagem , Ganglioneuroblastoma/tratamento farmacológico , Ganglioneuroblastoma/cirurgia , Humanos , Lactente , Recém-Nascido , Japão , Estimativa de Kaplan-Meier , Imageamento por Ressonância Magnética , Masculino , Neuroblastoma/diagnóstico por imagem , Neuroblastoma/tratamento farmacológico , Derrame Pleural/etiologia , Complicações Pós-Operatórias/etiologia , Intervalo Livre de Progressão , Estudos Prospectivos , Fatores de Risco , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X , Doenças Vasculares/etiologia , Vincristina/administração & dosagem , Vincristina/efeitos adversos
14.
Jpn J Clin Oncol ; 49(8): 743-748, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31053863

RESUMO

BACKGROUND: Our previous study reported a method for determining MYCN gene amplification (MNA) status using cell-free DNA in serum. We prospectively analyzed the serum MNA status using sera obtained before the initial diagnosis from patients with neuroblastoma and evaluated the utility of this method. METHODS: Eighty patients were enrolled in the study. The serum MYCN/NAGK ratio was assessed for all cases. RESULTS: Fifteen cases showed serum MNA, while 65 did not. Of the 80 total patients, tumor samples for a genetic analysis were not obtained from 27 due to the patients' condition or other reasons. For the 43 of 80 cases that had both serum and tumor samples analyzed, the serum-based MNA status matched to tumor-based MNA status (P < 0.001). The sensitivity and the specificity were 100%, respectively. Seven of 15 cases who diagnosed as MNA by serum-based MNA status were <18 months of age, and tumor samples were not obtained from 4 of these cases. Based on the serum MNA status, these cases were able to start treatment immediately. The 4-year event-free survival rates of cases with and without MNA in sera were 37.5% and 84.8%, respectively (P < 0.001). CONCLUSION: The serum-based MNA status was useful for precisely predicting the MNA status in tumor and it has clinical benefits for predicting risk stratification in patients for whom obtaining tumor samples is difficult.


Assuntos
Amplificação de Genes , Biópsia Líquida , Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/genética , Pré-Escolar , Intervalo Livre de Doença , Feminino , Humanos , Lactente , Masculino , Neuroblastoma/sangue , Neuroblastoma/diagnóstico , Neuroblastoma/cirurgia , Prognóstico , Estudos Prospectivos
15.
Endocr J ; 66(12): 1117-1123, 2019 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-31484850

RESUMO

A 14-year-old girl was referred to our department because of headache and visual impairment following the resection of recurrent cardiac myxoma. Head magnetic resonance imaging (MRI) scan detected an intra- and supra-sellar tumor. Moreover, the patient showed the presence of spotty skin pigmentations on her cheeks and lower lip. Blood examination revealed hypothyrotropinemia, and ultrasonography results revealed multiple thyroid nodules. She was diagnosed with Carney complex (CNC). Her pituitary tumor was suspected as growth hormone (GH)-secreting adenoma, because overgrowth was observed in the patient. However, biochemical examinations, including oral glucose tolerance test, failed to show the characteristic findings of GH-secreting adenoma. In contrast, insulin tolerance test showed GH deficiency. Her visual impairment improved without performing decompression surgery, and the tumor size decreased, as per the MRI findings. Based on clinical course, the patient was diagnosed with pituitary apoplexy in pituitary adenoma, following which she was discharged. At 3 months after discharge, thyrotropin-releasing hormone loading test performed revealed low thyrotropin-stimulating hormone and thyroid hormone levels, and the patient was in a depressed mood. Therefore, l-T4 replacement was initiated, following which her GH secretory capacity gradually improved. Here, we report, to the best of our knowledge, the first case of a patient with pituitary apoplexy in CNC. Such condition must be identified in young patients with recurrent cardiac myxoma, and examinations, such as head MRI, must be performed.


Assuntos
Procedimentos Cirúrgicos Cardíacos/efeitos adversos , Complexo de Carney/complicações , Neoplasias Cardíacas/cirurgia , Mixoma/cirurgia , Apoplexia Hipofisária/etiologia , Neoplasias Hipofisárias/complicações , Adolescente , Complexo de Carney/diagnóstico , Feminino , Adenoma Hipofisário Secretor de Hormônio do Crescimento/complicações , Humanos , Recidiva Local de Neoplasia/cirurgia , Neoplasias Hipofisárias/diagnóstico , Neoplasias Hipofisárias/cirurgia , Tireotropina/deficiência
16.
Blood ; 125(25): 3905-16, 2015 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-25940712

RESUMO

Although T cells expressing CD19-specific chimeric antigen receptors (CARs) are a promising new therapy for B-cell malignancies, objective responses are observed at lower frequencies in patients with lymphoma than in those with acute B-cell leukemia. We postulated that the tumor microenvironment suppresses CAR-expressing T cells (CARTs) through the activity of indoleamine 2,3-dioxygenase (IDO), an intracellular enzyme that converts tryptophan into metabolites that inhibit T -: cell activity. To investigate the effects of tumor IDO on CD19-CART therapy, we used a xenograft lymphoma model expressing IDO as a transgene. CD19-CARTs inhibited IDO-negative tumor growth but had no effect on IDO-positive tumors. An IDO inhibitor (1-methyl-tryptophan) restored IDO-positive tumor control. Moreover, tryptophan metabolites inhibited interleukin (IL)-2-, IL-7-, and IL-15-dependent expansion of CARTs; diminished their proliferation, cytotoxicity, and cytokine secretion in vitro in response to CD19 recognition; and increased their apoptosis. Inhibition of CD19-CARTs was not mitigated by the incorporation of costimulatory domains, such as 4-1BB, into the CD19-CAR. Finally, we found that fludarabine and cyclophosphamide, frequently used before CART administration, downregulated IDO expression in lymphoma cells and improved the antitumor activity of CD19-CART in vivo. Because tumor IDO inhibits CD19-CARTs, antagonizing this enzyme may benefit CD19-CART therapy.


Assuntos
Antígenos CD19/imunologia , Imunoterapia/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Linfoma/enzimologia , Linfoma/imunologia , Linfócitos T/imunologia , Animais , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Ciclofosfamida/farmacologia , Modelos Animais de Doenças , Regulação para Baixo , Citometria de Fluxo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/efeitos dos fármacos , Camundongos , Camundongos SCID , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Antígenos de Linfócitos T/imunologia , Proteínas Recombinantes de Fusão , Linfócitos T/efeitos dos fármacos , Vidarabina/análogos & derivados , Vidarabina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Jpn J Clin Oncol ; 46(7): 661-6, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27207883

RESUMO

BACKGROUND: It remains unclear whether a residual tumor mass following therapy influences the prognosis of neuroblastoma. METHODS: We retrospectively reviewed 20 patients with intermediate-risk tumors treated at our institution between 1993 and 2012 to elucidate whether additional treatment is required for residual tumors. RESULTS: The patient ages at diagnosis ranged from 0 days to 7 years. The 5-year overall survival rate was 94.4%. Thirteen patients had Stage 3 disease and seven patients had Stage 4 disease. Nine patients showed intraspinal extension. Twelve patients had a residual tumor mass at the completion of therapy, and eight showed intraspinal extension. Five of these 12 patients showed metaiodobenzylguanidine (MIBG) uptake at the end of treatment, but the uptake disappeared during the follow-up period. Except for one patient who died due to treatment complications, the rest are all alive, and nine are alive with a residual mass. We examined the residual mass in four patients and found that these tissues had differentiated into a ganglioneuroma or changed to a necrotic tissue. For the three patients with neurological symptoms at the end of treatment, some slight neurological symptoms still remained during the follow-up. Five patients with an intraspinal mass eventually presented with new symptoms. CONCLUSIONS: The presence of a residual mass at the end of treatment did not influence the patients' prognosis. Therefore, an invasive radical surgical resection and additional treatment may not be necessary. Cases with a residual intraspinal mass also require a long-term follow-up to assess the neurological prognosis.The presence of a residual mass in cases of intermediate-risk neuroblastoma at the end of treatment did not influence the patients' prognosis.


Assuntos
Neoplasia Residual/mortalidade , Neoplasia Residual/patologia , Neuroblastoma/mortalidade , Neuroblastoma/patologia , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Neoplasia Residual/terapia , Neuroblastoma/terapia , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento
18.
Mol Ther ; 23(9): 1475-85, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26022733

RESUMO

Human induced pluripotent stem cells (hiPSC) hold promise for regenerative therapies, though there are several safety concerns including the risk of oncogenic transformation or unwanted adverse effects associated with hiPSC or their differentiated progeny. Introduction of the inducible caspase-9 (iC9) suicide gene, which is activated by a specific chemical inducer of dimerization (CID), is one of the most appealing safety strategies for cell therapies and is currently being tested in multicenter clinical trials. Here, we show that the iC9 suicide gene with a human EF1α promoter can be introduced into hiPSC by lentiviral transduction. The transduced hiPSC maintain their pluripotency, including their capacity for unlimited self-renewal and the potential to differentiate into three germ layer tissues. Transduced hiPSC are eliminated within 24 hours of exposure to pharmacological levels of CID in vitro, with induction of apoptosis in 94-99% of the cells. Importantly, the iC9 suicide gene can eradicate tumors derived from hiPSC in vivo. In conclusion, we have developed a direct and efficient hiPSC killing system that provides a necessary safety mechanism for therapies using hiPSC. We believe that our iC9 suicide gene will be of value in clinical applications of hiPSC-based therapy.


Assuntos
Caspase 9/genética , Expressão Gênica , Genes Transgênicos Suicidas , Terapia Genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Caspase 9/metabolismo , Diferenciação Celular , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ordem dos Genes , Genes Reporter , Terapia Genética/métodos , Vetores Genéticos/genética , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Lentivirus/genética , Masculino , Camundongos , Teratoma/genética , Teratoma/patologia , Teratoma/terapia , Transgenes , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Mol Ther ; 23(9): 1497-506, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26084970

RESUMO

Oncolytic adenoviruses (OAdV) represent a promising strategy for cancer therapy. Despite their activity in preclinical models, to date the clinical efficacy remains confined to minor responses after intratumor injection. To overcome these limitations, we developed an alternative approach using the combination of the OAdv ICOVIR15 with a replication incompetent adenoviral vector carrying the suicide gene of inducible Caspase 9 (Ad.iC9), both of which are delivered by mesenchymal stromal cells (MSCs). We hypothesized that coinfection with ICOVIR15 and Ad.iC9 would allow MSCs to replicate both vectors and deliver two distinct types of antitumor therapy to the tumor, amplifying the cytotoxic effects of the two viruses, in a non-small-cell lung cancer (NSCLC) model. We showed that MSCs can replicate and release both vectors, enabling significant transduction of the iC9 gene in tumor cells. In the in vivo model using human NSCLC xenografts, MSCs homed to lung tumors where they released both viruses. The activation of iC9 by the chemical inducer of dimerization (CID) significantly enhanced the antitumor activity of the ICOVIR15, increasing the tumor control and translating into improved overall survival of tumor-bearing mice. These data support the use of this innovative approach for the treatment of NSCLC.


Assuntos
Adenoviridae/genética , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Terapia Genética , Vetores Genéticos/genética , Neoplasias Pulmonares/genética , Células-Tronco Mesenquimais/metabolismo , Terapia Viral Oncolítica , Animais , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/terapia , Caspase 9/genética , Caspase 9/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Ativação Enzimática , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Genes Reporter , Genes Transgênicos Suicidas , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intralesionais , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Transplante de Células-Tronco Mesenquimais , Camundongos , Terapia Viral Oncolítica/métodos , Proteínas Recombinantes de Fusão/genética , Proteína 1A de Ligação a Tacrolimo/genética , Transdução Genética , Transgenes , Replicação Viral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Rinsho Ketsueki ; 56(4): 428-31, 2015 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-25971275

RESUMO

A 14-year-old male with multiple myeloma (IgG-λ, ISS stage 3) received myeloablative matched unrelated bone marrow transplantation, and achieved a complete response. At 16 months after the transplantation, he relapsed. The relapse was resistant to bortezomib and thalidomide. Peripheral blood showed mixed chimerism with 10% recipient cells. Peripheral blood stem cells (PBSC) were collected and pseudo-autologous PBSC transplantation (PASCT) was performed following high-dose melphalan without graft-versus-host disease prophylaxis. Hematopoietic recovery was prompt and a partial response was obtained without graft-versus-host disease exacerbation. We have presented a rare case of adolescent-onset multiple myeloma, obtaining a transient response with PASCT following post-allogeneic transplant relapse.


Assuntos
Transplante de Medula Óssea , Doença Enxerto-Hospedeiro , Mieloma Múltiplo/terapia , Recidiva Local de Neoplasia/terapia , Adolescente , Transplante de Medula Óssea/métodos , Humanos , Masculino , Mieloma Múltiplo/diagnóstico , Recidiva Local de Neoplasia/diagnóstico , Recidiva , Condicionamento Pré-Transplante/métodos , Transplante Homólogo/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA