Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Cancer ; 143(11): 2838-2848, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30144023

RESUMO

Demethylation of the long interspersed nuclear element (LINE-1; L1) antisense promoter can result in transcription of neighboring sequences as for the L1-MET transcript produced by the L1 placed in the second intron of MET. To define the role of L1-MET, we investigated the sequence and the transcription of L1-MET in vitro models and heterogeneous breast cancers, previously reported to show other L1-derived transcripts. L1-MET expressing cell lines were initially identified in silico and investigated for L1-MET promoter methylation, cDNA sequence and cell fraction mRNA. The transcriptional level of L1-MET and MET were then evaluated in breast specimens, including 9 cancer cell lines, 41 carcinomas of different subtypes, and 11 normal tissues. In addition to a L1-MET transcript ending at MET exon 21, six novel L1-MET splice variants were identified. Normal breast tissues were negative for the L1-MET expression, whereas the triple-negative breast cancer (TNBC) and the high-grade carcinomas were enriched with the L1-MET mRNA (p = 0.005 and p = 0.018, respectively). In cancer cells and tissues the L1-MET expression was associated with its promoter hypomethylation (ρ = -0.8 and -0.9, respectively). No correlation was found between L1-MET and MET mRNA although L1-MET expressing tumors with higher L1-MET/MET ratio were negative for the MET protein expression (p = 0.006). Besides providing the first identification and detailed description of L1-MET in breast cancer, we clearly demonstrate that higher levels of this transcript specifically recognize a subset of more aggressive carcinomas, mainly TNBC. We suggest the possible evaluation of L1-MET in the challenging diagnosis of early TNBCs.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Proteínas Proto-Oncogênicas c-met/genética , Neoplasias de Mama Triplo Negativas/genética , Células A549 , Mama/metabolismo , Linhagem Celular Tumoral , Metilação de DNA/genética , Feminino , Células HCT116 , Humanos , Regiões Promotoras Genéticas/genética , Splicing de RNA/genética , RNA Mensageiro/genética
2.
Mol Cancer ; 16(1): 86, 2017 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-28454547

RESUMO

BACKGROUND: Enhancing the antitumor activity of the DNA-damaging drugs is an attractive strategy to improve current treatment options. Trabectedin is an isoquinoline alkylating agent with a peculiar mechanism of action. It binds to minor groove of DNA inducing single- and double-strand-breaks. These kinds of damage lead to the activation of PARP1, a first-line enzyme in DNA-damage response pathways. We hypothesized that PARP1 targeting could perpetuate trabectedin-induced DNA damage in tumor cells leading finally to cell death. METHODS: We investigated trabectedin and PARP1 inhibitor synergism in several tumor histotypes both in vitro and in vivo (subcutaneous and orthotopic tumor xenografts in mice). We searched for key determinants of drug synergism by comparative genomic hybridization (aCGH) and gene expression profiling (GEP) and validated their functional role. RESULTS: Trabectedin activated PARP1 enzyme and the combination with PARP1 inhibitors potentiated DNA damage, cell cycle arrest at G2/M checkpoint and apoptosis, if compared to single agents. Olaparib was the most active PARP1 inhibitor to combine with trabectedin and we confirmed the antitumor and antimetastatic activity of trabectedin/olaparib combination in mice models. However, we observed different degree of trabectedin/olaparib synergism among different cell lines. Namely, in DMR leiomyosarcoma models the combination was significantly more active than single agents, while in SJSA-1 osteosarcoma models no further advantage was obtained if compared to trabectedin alone. aCGH and GEP revealed that key components of DNA-repair pathways were involved in trabectedin/olaparib synergism. In particular, PARP1 expression dictated the degree of the synergism. Indeed, trabectedin/olaparib synergism was increased after PARP1 overexpression and reduced after PARP1 silencing. CONCLUSIONS: PARP1 inhibition potentiated trabectedin activity in a PARP1-dependent manner and PARP1 expression in tumor cells might be a useful predictive biomarker that deserves clinical evaluation.


Assuntos
Biomarcadores Tumorais/genética , Dioxóis/administração & dosagem , Poli(ADP-Ribose) Polimerase-1/genética , Sarcoma/tratamento farmacológico , Tetra-Hidroisoquinolinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Hibridização Genômica Comparativa , Dano ao DNA/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Sarcoma/genética , Sarcoma/patologia , Trabectedina , Ensaios Antitumorais Modelo de Xenoenxerto
3.
J Neurooncol ; 131(2): 213-222, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27796734

RESUMO

The diagnosis of 206 low and high grade adult gliomas, including 40 oligoastrocytomas, was revised based on the immunohistochemical reactivity for the ATRX protein, IDH1/2 mutation status and 1p/19q chromosomal status. All oligodendrogliomas kept the initial diagnosis. Astrocytomas did not change diagnosis in 30 of 36 cases (83.3 %); four of 36 (11.1 %) cases were reclassified as oligodendroglioma, one (2.8 %) as DNT and the other (2.8 %) as reactive gliosis. Oligoastrocytomas changed diagnosis in 35 of 40 (87.5 %) cases, being reclassified 22 of 40 (55 %) as astrocytoma, 11 of 40 (27.5 %) as oligodendroglioma and two of 40 (5 %) as reactive gliosis. Four (10 %) remained unclassifiable. In one case only (2.5 %), the diagnosis of oligoastrocytoma could not be excluded since tumor astrocytes and tumor oligodendrocytes coexisted in mixed tumor areas. In the GBM tumor subgroup, GBMO disappeared because they were not substantiated by molecular genetics. Pilocytic astrocytomas retained ATRX expression. Loss of nuclear ATRX protein expression was strongly associated to IDH1/2 mutations (p = 0.0001) and mutually exclusive with total 1p/19q co-deletion (p = 0.0001). In astrocytic tumors, loss of immunoreactivity for the ATRX protein was significantly associated to the ALT phenotype (p = 0.0003). The constitutive ATRX expression in microglia/macrophages may be misleading, especially in the identification of an oligodendroglial tumor infiltration. Of paramount importance in the recognition of oligodendroglial and astrocytic tumor cells were the double immunostainings for ATRX/GFAP, ATRX/IDH1R132H, ATRX/Iba-1 and ATRX/CD68.


Assuntos
Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Proteína Nuclear Ligada ao X/metabolismo , Astrocitoma/diagnóstico , Astrocitoma/metabolismo , Astrocitoma/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Deleção Cromossômica , Cromossomos Humanos Par 1/genética , Cromossomos Humanos Par 19/genética , Diagnóstico Diferencial , Deleção de Genes , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Humanos , Isocitrato Desidrogenase/metabolismo , Macrófagos/metabolismo , Microglia/metabolismo , Oligodendroglioma/diagnóstico , Oligodendroglioma/metabolismo , Oligodendroglioma/patologia
4.
Tumour Biol ; 37(3): 4041-52, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26486326

RESUMO

Biliary tract carcinoma is a rare malignancy with multiple causes, which underlie the different genetic and molecular profiles. Cancer cell lines are affordable models, reflecting the characteristics of the tumor of origin. They represent useful tools to identify molecular targets for treatment. Here, we established and characterized from biological, molecular, and genetic point of view, an Italian intrahepatic cholangiocarcinoma cell line (ICC), the MT-CHC01. MT-CHC01 cells were isolated from a tumor-derived xenograft. Immunophenotypical characterization was evaluated both at early and after stabilization passages. In vitro biological, genetic, and molecular features were also investigated. In vivo tumorigenicity was assessed in NOD/SCID mice. MT-CHC01cells retain epithelial cell markers, EPCAM, CK7, and CK19, and some stemness and pluripotency markers, i.e., SOX2, Nanog, CD49f/integrin-α6, CD24, PDX1, FOXA2, and CD133. They grow as a monolayer, with a population double time of about 40 h; they show a low migration and invasion potential. In low attachment conditions, they are able to form spheres and to growth in anchorage-independent manner. After subcutaneous injection, they retain in vivo tumorigenicity; the expression of biliary markers as CA19-9 and CEA were maintained from primary tumor. The karyotype is highly complex, with a hypotriploid to hypertriploid modal number (3n+/-) (52 to 77 chromosomes); low level of HER2 gene amplification, TP53 deletion, gain of AURKA were identified; K-RAS G12D mutation were maintained from primary tumor to MT-CHC01 cells. We established the first ICC cell line derived from an Italian patient. It will help to study either the biology of this tumor or to test drugs both in vitro and in vivo.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Linhagem Celular Tumoral/fisiologia , Colangiocarcinoma/patologia , Animais , Neoplasias dos Ductos Biliares/genética , Testes de Carcinogenicidade , Movimento Celular , Colangiocarcinoma/genética , Hibridização Genômica Comparativa , Análise Mutacional de DNA , Feminino , Humanos , Itália , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Transplante de Neoplasias , Proteínas Proto-Oncogênicas p21(ras)/genética
5.
BMC Cancer ; 15: 374, 2015 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-25952930

RESUMO

BACKGROUND: Malignant Pleural Mesothelioma (MPM) is an aggressive tumor arising from mesothelial cells lining the pleural cavities characterized by resistance to standard therapies. Most of the molecular steps responsible for pleural transformation remain unclear; however, several growth factor signaling cascades are known to be altered during MPM onset and progression. Transducers of these pathways, such as PIK3CA-mTOR-AKT, MAPK, and ezrin/radixin/moesin (ERM) could therefore be exploited as possible targets for pharmacological intervention. This study aimed to identify 'druggable' pathways in MPM and to formulate a targeted approach based on the use of commercially available molecules, such as the multikinase inhibitor sorafenib and the mTOR inhibitor everolimus. METHODS: We planned a triple approach based on: i) analysis of immunophenotypes and mutational profiles in a cohort of thoracoscopic MPM samples, ii) in vitro pharmacological assays, ii) in vivo therapeutic approaches on MPM xenografts. No mutations were found in 'hot spot' regions of the mTOR upstream genes (e.g. EGFR, KRAS and PIK3CA). RESULTS: Phosphorylated mTOR and ERM were specifically overexpressed in the analyzed MPM samples. Sorafenib and everolimus combination was effective in mTOR and ERM blockade; exerted synergistic effects on the inhibition of MPM cell proliferation; triggered ROS production and consequent AMPK-p38 mediated-apoptosis. The antitumor activity was displayed when orally administered to MPM-bearing NOD/SCID mice. CONCLUSIONS: ERM and mTOR pathways are activated in MPM and 'druggable' by a combination of sorafenib and everolimus. Combination therapy is a promising therapeutic strategy against MPM.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Proteínas do Citoesqueleto/metabolismo , Everolimo/administração & dosagem , Mesotelioma/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/administração & dosagem , Neoplasias Pleurais/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo , Administração Oral , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Proteínas do Citoesqueleto/genética , Sinergismo Farmacológico , Everolimo/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Mesotelioma/genética , Mesotelioma/metabolismo , Camundongos , Niacinamida/administração & dosagem , Niacinamida/farmacologia , Compostos de Fenilureia/farmacologia , Fosforilação/efeitos dos fármacos , Neoplasias Pleurais/genética , Neoplasias Pleurais/metabolismo , Sorafenibe , Serina-Treonina Quinases TOR/genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Rep ; 36(4): 109455, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34320350

RESUMO

In glioblastoma (GBM), the most frequent and lethal brain tumor, therapies suppressing recurrently altered signaling pathways failed to extend survival. However, in patient subsets, specific genetic lesions can confer sensitivity to targeted agents. By exploiting an integrated model based on patient-derived stem-like cells, faithfully recapitulating the original GBMs in vitro and in vivo, here, we identify a human GBM subset (∼9% of all GBMs) characterized by ERBB3 overexpression and nuclear accumulation. ERBB3 overexpression is driven by inheritable promoter methylation or post-transcriptional silencing of the oncosuppressor miR-205 and sustains the malignant phenotype. Overexpressed ERBB3 behaves as a specific signaling platform for fibroblast growth factor receptor (FGFR), driving PI3K/AKT/mTOR pathway hyperactivation, and overall metabolic upregulation. As a result, ERBB3 inhibition by specific antibodies is lethal for GBM stem-like cells and xenotransplants. These findings highlight a subset of patients eligible for ERBB3-targeted therapy.


Assuntos
Glioblastoma/genética , MicroRNAs/metabolismo , Receptor ErbB-3/metabolismo , Anticorpos/metabolismo , Apoptose , Linhagem Celular Tumoral , Fator 2 de Crescimento de Fibroblastos , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , MicroRNAs/genética , Oligodendroglia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Prognóstico , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-3/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Esferoides Celulares/patologia , Serina-Treonina Quinases TOR/metabolismo
7.
Am J Surg Pathol ; 42(9): 1190-1200, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29975246

RESUMO

The American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) 2013 guidelines for HER2 assessment have increased the number of HER2 equivocal breast carcinomas following in situ hybridization reflex testing, that is, HER2 "double equivocal" (equivocal protein expression and equivocal gene copy number). Forty-five double-equivocal carcinomas were subjected to Prosigna analysis. Twenty-seven cases were investigated for the expression of genes found to be differentially expressed between estrogen receptor (ER)-positive/HER2-positive (N=22) and ER-positive/HER2-negative (N=22) control cases. Twenty-nine of the 45 cases were also analyzed by targeted sequencing using a panel of 14 genes. We then explored the pathologic complete response rates in an independent series of double-equivocal carcinoma patients treated with trastuzumab-containing chemotherapy. All cases were ER-positive, with a mean Ki67 of 28%. Double-equivocal carcinomas were predominantly luminal B (76%); 9 cases (20%) were luminal A, and 2 cases (4%) HER2-enriched. The majority (73%) showed a high risk of recurrence by Prosigna, even when the carcinomas were small (<2 cm), node-negative/micrometastatic, and/or grade 2. Double-equivocal carcinomas showed TP53 (6/29, 20%), PIK3CA (3/29, 10%), HER2 (1/29, 3%), and MAP2K4 (1/29, 3%) mutations. Compared with grade-matched ER-positive/HER2-negative breast carcinomas from METABRIC, double-equivocal carcinomas harbored more frequently TP53 mutations and less frequently PIK3CA mutations (P<0.05). No significant differences were observed with grade-matched ER-positive/HER2-positive carcinomas. Lower pathologic complete response rates were observed in double-equivocal compared with HER2-positive patients (10% vs. 60%, P=0.009). Double-equivocal carcinomas are preferentially luminal B and show a high risk of recurrence. A subset of these tumors can be labeled as HER2-enriched by transcriptomic analysis. HER2 mutations can be identified in HER2 double-equivocal cases.


Assuntos
Biomarcadores Tumorais/análise , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma/genética , Carcinoma/patologia , Receptor ErbB-2/genética , Adulto , Biomarcadores Tumorais/genética , Feminino , Perfilação da Expressão Gênica , Humanos , Pessoa de Meia-Idade
8.
Hum Pathol ; 46(3): 411-8, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25596657

RESUMO

A crucial role for Aurora Kinase A (AURKA) gene has been demonstrated in the advanced steps of colorectal tumor progression. Little is known, however, about its role in the early phases of the adenoma-carcinoma sequence. Moreover, no data are currently available concerning AURKA involvement in the serrated tumorigenesis. Fluorescence in situ hybridization analysis and immunohistochemistry were used to assess gene copy number and protein expression in 40 colorectal adenomas, 20 cancerized adenomas, and 20 serrated polyps. An increased copy number was found either in adenomatous tissue or in early cancer in the vast majority of cancerized adenomas, but only in 5% of adenomas (P < .001). Protein expression strictly paralleled fluorescence in situ hybridization results. No changes in the gene copy number were observed in serrated polyps, regardless of their histotype and the presence of dysplasia, even if high percentages of immunostained cells were detected in all the subgroups. AURKA gene is associated with progressive colorectal adenomas but is uninvolved in the development of nonprogressive adenomas. The diploid status of the gene is maintained along the progression of serrated neoplasia. AURKA protein expression in serrated polyps is uncoupled from gene status and is likely to reflect apoptotic dysregulation.


Assuntos
Adenoma/enzimologia , Adenoma/genética , Aurora Quinase A/genética , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/genética , Dosagem de Genes/genética , Adenoma/patologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Pólipos do Colo/enzimologia , Pólipos do Colo/genética , Pólipos do Colo/patologia , Neoplasias Colorretais/patologia , Progressão da Doença , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Gradação de Tumores , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia
9.
Cancer Res ; 74(1): 119-29, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24356422

RESUMO

Unresectable metastatic bone sarcoma and soft-tissue sarcomas (STS) are incurable due to the inability to eradicate chemoresistant cancer stem-like cells (sCSC) that are likely responsible for relapses and drug resistance. In this study, we investigated the preclinical activity of patient-derived cytokine-induced killer (CIK) cells against autologous bone sarcoma and STS, including against putative sCSCs. Tumor killing was evaluated both in vitro and within an immunodeficient mouse model of autologous sarcoma. To identify putative sCSCs, autologous bone sarcoma and STS cells were engineered with a CSC detector vector encoding eGFP under the control of the human promoter for OCT4, a stem cell gene activated in putative sCSCs. Using CIK cells expanded from 21 patients, we found that CIK cells efficiently killed allogeneic and autologous sarcoma cells in vitro. Intravenous infusion of CIK cells delayed autologous tumor growth in immunodeficient mice. Further in vivo analyses established that CIK cells could infiltrate tumors and that tumor growth inhibition occurred without an enrichment of sCSCs relative to control-treated animals. These results provide preclinical proof-of-concept for an effective strategy to attack autologous sarcomas, including putative sCSCs, supporting the clinical development of CIK cells as a novel class of immunotherapy for use in settings of untreatable metastatic disease.


Assuntos
Células Matadoras Induzidas por Citocinas/imunologia , Citocinas/farmacologia , Imunoterapia Adotiva/métodos , Sarcoma/imunologia , Sarcoma/terapia , Animais , Carcinogênese/imunologia , Carcinogênese/patologia , Linhagem Celular Tumoral , Células Matadoras Induzidas por Citocinas/efeitos dos fármacos , Citocinas/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Sarcoma/patologia
10.
Clin Cancer Res ; 19(8): 2117-31, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23434734

RESUMO

PURPOSE: The multikinase inhibitor sorafenib displays antitumor activity in preclinical models of osteosarcoma. However, in sorafenib-treated patients with metastatic-relapsed osteosarcoma, disease stabilization and tumor shrinkage were short-lived and drug resistance occurred. We explored the sorafenib treatment escape mechanisms to overcome their drawbacks. EXPERIMENTAL DESIGN: Immunoprecipitation, Western blotting, and immunohistochemistry were used to analyze the mTOR pathway [mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2)]. Cell viability, colony growth, and cell migration were evaluated in different osteosarcoma cell lines (MNNG-HOS, HOS, KHOS/NP, MG63, U-2OS, SJSA-1, and SAOS-2) after scalar dose treatment with sorafenib (10-0.625 µmol/L), rapamycin-analog everolimus (100-6.25 nmol/L), and combinations of the two. Cell cycle, reactive oxygen species (ROS) production, and apoptosis were assessed by flow cytometry. Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice injected with MNNG-HOS cells were used to determine antitumor and antimetastatic effects. Angiogenesis and vascularization were evaluated in vitro by exploiting endothelial branching morphogenesis assays and in vivo in xenografted mice and chorioallantoic membranes. RESULTS: After sorafenib treatment, mTORC1 signaling was reduced (downstream target P-S6), whereas mTORC2 was increased (phospho-mTOR Ser2481) in MNNG-HOS xenografts compared with vehicle-treated mice. Combining sorafenib with everolimus resulted in complete abrogation of both mTORC1 [through ROS-mediated AMP-activated kinase (AMPK) activation] and mTORC2 (through complex disassembly). The sorafenib/everolimus combination yielded: (i) enhanced antiproliferative and proapoptotic effects, (ii) impaired tumor growth, (iii) potentiated antiangiogenesis, and (iv) reduced migratory and metastatic potential. CONCLUSION: mTORC2 activation is an escape mechanism from sorafenib treatment. When sorafenib is combined with everolimus, its antitumor activity is increased by complete inhibition of the mTOR pathway in the preclinical setting.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Complexos Multiproteicos/metabolismo , Osteossarcoma/tratamento farmacológico , Serina-Treonina Quinases TOR/metabolismo , Regulação para Cima/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Embrião de Galinha , Everolimo , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Patológica/metabolismo , Neovascularização Patológica/prevenção & controle , Niacinamida/administração & dosagem , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Osteossarcoma/irrigação sanguínea , Osteossarcoma/patologia , Compostos de Fenilureia/administração & dosagem , Compostos de Fenilureia/farmacologia , Interferência de RNA , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/administração & dosagem , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Sorafenibe
11.
J Immunother ; 35(7): 579-86, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22892454

RESUMO

Cytokine-induced killer cells (CIKs) are ex vivo expanded T-NK lymphocytes capable of HLA-unrestricted antitumor activity. CIKs are promising candidates for adoptive cancer immunotherapies; they can be generated and infused in autologous settings of cancer patients, or from donors, after allogeneic hematopoietic cell transplant. Ex vivo expansion rates of CIKs are greatly variable among patients, with consequent potential clinical limitations for "poor expanders." We compared the standard expansion protocol with a new one, which included the timed addition of irradiated allogeneic peripheral blood mononuclear cells. Our hypothesis is that allogeneic stimulation might provide CIK cells with a proliferative boost and simultaneously decrease their alloreactivity versus third parties, if HLA-mismatched from the allogeneic stimulators. Allo-stimulated CIKs (AS-CIK) reached significantly higher expansion rates compared with standard controls, regardless if generated form healthy donors (131- vs. 32-fold) or cancer patients (117- vs. 14-fold). The expansion of the CD3CD56 subset was 2243-fold for AS-CIKs compared with 362 for standard CIKs. AS-CIKs efficiently killed osteosarcoma targets in vitro, results were comparable with that of standard CIKs. Standard and AS-CIKs did not show differences in phenotype and telomere length. The alloreactivity of AS-CIKs against third party HLA-mismatched peripheral blood mononuclear cells was reduced compared with standard CIKs (37% vs. 23%). In conclusion, alloreactivity of CIK cells may be exploited enhancing their final ex vivo expansion. In clinical perspective these findings may facilitate the extension of CIK-based immunotherapy to larger numbers of patients and, translated into hematopoietic cell transplant settings, contribute to reduce the risk of graft versus host disease in the hypothesis of infusions across HLA barriers.


Assuntos
Neoplasias Ósseas/terapia , Células Matadoras Induzidas por Citocinas/imunologia , Imunoterapia Adotiva , Osteossarcoma/terapia , Neoplasias Ósseas/imunologia , Neoplasias Ósseas/patologia , Complexo CD3/metabolismo , Antígeno CD56/metabolismo , Técnicas de Cultura de Células/métodos , Proliferação de Células , Células Cultivadas , Células Matadoras Induzidas por Citocinas/transplante , Citotoxicidade Imunológica , Antígenos HLA/imunologia , Humanos , Imunização , Isoantígenos/imunologia , Leucócitos Mononucleares/imunologia , Osteossarcoma/imunologia , Osteossarcoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA