Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Histopathology ; 80(4): 686-697, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34821406

RESUMEN

AIMS: To evaluate the diagnostic accuracy of SSX and SSX::SS18 antibodies in decalcified surgical specimens and outcome of synovial sarcomas (SS) of bone. METHODS AND RESULTS: Twenty-five cases were classified as bone SS (prevalence 0.32% among malignant primary bone sarcoma). Median age was 34 years (range = 9-79). Twenty-four of 25 patients presented with non-metastatic tumours, one with lung metastases. The majority of tumours involved the long bones of extremities with metaphyseal origin. Mean size of the tumour was 7.1 cm. Twenty cases (80%) were monophasic and five (20%) were biphasic. SS18::SSX fusion-specific antibody had 92% sensitivity and 99% specificity for primary bone SS, whereas SSX C-terminus antibody had 100% sensitivity and 94% specificity. Fluorescence in-situ hybridisation (FISH) analysis was feasible in nine (36%) cases and detected SS18 rearrangement in all nine cases. All patients underwent surgical removal of their primary tumour, with adequate margins in 18 (72%) patients. Chemotherapy with metothrexate, cisplatin, doxorubicin and ifosfamide was used in the seven patients. Two patients with inadequate surgical margins received radiotherapy. With a median follow-up of 80 months (range = 6-428), 5- and 10-year overall survival (OS) was 66.6% and 47.9%, respectively, and 5 and 10 years' disease-free survival (DFS) was 36.8% [95% confidence interval (CI) = 18.0-55.7%] and 32.2% (95% CI = 14.6-51.2%), respectively. A significant improvement in 10 years' DFS in patients undergoing chemotherapy compared with patients who did not was observed (P = 0.039). CONCLUSIONS: Our series highlights the utility of SS18::SSX fusion-specific and SSX C-terminus antibodies to support the diagnosis of SS. Adjustment chemotherapy was associated with improved prognosis in this series.


Asunto(s)
Anticuerpos/análisis , Neoplasias Óseas/diagnóstico , Proteínas de Fusión Oncogénica/inmunología , Sarcoma Sinovial/diagnóstico , Adolescente , Adulto , Anciano , Neoplasias Óseas/patología , Niño , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Sarcoma Sinovial/patología , Adulto Joven
2.
Mod Pathol ; 34(4): 748-757, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33299109

RESUMEN

Alveolar Rhabdomyosarcoma (ARMS) is an aggressive pediatric cancer with about 80% of cases characterized by either a t(1;13)(p36;q14) or t(2;13)(q35;q14), which results in the formation of the fusion oncogenes PAX7-FOXO1 and PAX3-FOXO1, respectively. Since patients with fusion-positive ARMS (FP-RMS) have a poor prognosis and are treated with an aggressive therapeutic regimen, correct classification is of clinical importance. Detection of the translocation by different molecular methods is used for diagnostics, including fluorescence in situ hybridization and RT-PCR or NGS based approaches. Since these methods are complex and time consuming, we developed specific monoclonal antibodies (mAbs) directed to the junction region on the PAX3-FOXO1 fusion protein. Two mAbs, PFM.1 and PFM.2, were developed and able to immunoprecipitate in vitro-translated PAX3-FOXO1 and cellular PAX3-FOXO1 from FP-RMS cells. Furthermore, the mAbs recognized a 105 kDa band in PAX3-FOXO1-transfected cells and in FP-RMS cell lines. The mAbs did not recognize proteins in fusion-negative embryonal rhabdomyosarcoma cell lines, nor did they recognize PAX3 or FOXO1 alone when compared to anti-PAX3 and anti-FOXO1 antibodies. We next evaluated the ability of mAb PFM.2 to detect the fusion protein by immunohistochemistry. Both PAX3-FOXO1 and PAX7-FOXO1 were detected in HEK293 cells transfected with the corresponding cDNAs. Subsequently, we stained 26 primary tumor sections and a rhabdomyosarcoma tissue array and detected both fusion proteins with a positive predictive value of 100%, negative predictive value of 98%, specificity of 100% and a sensitivity of 91%. While tumors are stained homogenously in PAX3-FOXO1 cases, the staining pattern is heterogenous with scattered positive cells only in tumors expressing PAX7-FOXO1. No staining was observed in stromal cells, embryonal rhabdomyosarcoma, and fusion-negative rhabdomyosarcoma. These results demonstrate that mAbs specific for the chimeric oncoproteins PAX3-FOXO1 and PAX7-FOXO1 can be used efficiently for simple and fast subclassification of rhabdomyosarcoma in routine diagnostics via immunohistochemical detection.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Biomarcadores de Tumor/análisis , Inmunohistoquímica , Proteínas de Fusión Oncogénica/análisis , Factores de Transcripción Paired Box/análisis , Rabdomiosarcoma Alveolar/inmunología , Adolescente , Adulto , Animales , Especificidad de Anticuerpos , Niño , Preescolar , Femenino , Células HEK293 , Células HeLa , Humanos , Lactante , Masculino , Ratones , Persona de Mediana Edad , Células 3T3 NIH , Proteínas de Fusión Oncogénica/inmunología , Factores de Transcripción Paired Box/inmunología , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Rabdomiosarcoma Alveolar/patología , Adulto Joven
3.
Am J Hematol ; 94(3): 338-345, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30575108

RESUMEN

The prevalence, clinicopathologic correlates, and outcomes of previously untreated chronic lymphocytic leukemia (CLL) patients with IGH-BCL2 and IGH-BCL3 translocations are not well known. Using the Mayo Clinic CLL database, we identified patients seen between March 1, 2002 and September 30, 2016 who had FISH testing performed within 3 years of CLL diagnosis. The prognostic profile, time to first therapy (TTT), and overall survival (OS) of patients with IGH-BCL2 and IGH-BCL3 translocation were compared to patients without these abnormalities (non-IGH group). Of 1684 patients who met the inclusion criteria, 38 (2.2%) had IGH-BCL2, and 16 (0.9%) had IGH-BCL3 translocation at diagnosis. Patients with IGH-BCL3 translocation were more likely to have high and very-high CLL-International Prognostic Index, compared to patients with IGH-BCL2 translocation and the non-IGH group. The 5-year probability of requiring therapy was significantly higher for IGH-BCL3 compared to IGH-BCL2 and non-IGH groups (84% vs 33% vs 29%, respectively, P < 0.0001). The 5-year OS was significantly shorter for IGH-BCL3 compared to IGH-BCL2 and non-IGH groups (45% vs 89% vs 86%, respectively, P < 0.0001). On multivariable analyses, IGH-BCL3 translocation was associated with a shorter TTT (hazard ratio [HR] = 2.7; P = 0.005) and shorter OS (HR = 5.5; P < 0.0001); IGH-BCL2 translocation did not impact TTT and OS. In conclusion, approximately 3% of all newly diagnosed CLL patients have either an IGH-BCL2 or IGH-BCL3 translocation. Patients with IGH-BCL3 translocations have a distinct prognostic profile and outcome. These results support the inclusion of an IGH probe during the routine evaluation of FISH abnormalities in newly diagnosed CLL.


Asunto(s)
Proteínas del Linfoma 3 de Células B/genética , Cadenas Pesadas de Inmunoglobulina/genética , Leucemia Linfocítica Crónica de Células B/genética , Proteínas de Fusión Oncogénica/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Adenina/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Antraciclinas/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Proteínas del Linfoma 3 de Células B/inmunología , Estudios de Cohortes , Femenino , Expresión Génica , Humanos , Cadenas Pesadas de Inmunoglobulina/inmunología , Leucemia Linfocítica Crónica de Células B/diagnóstico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/mortalidad , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/inmunología , Piperidinas , Pronóstico , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Pirazoles/uso terapéutico , Pirimidinas/uso terapéutico , Análisis de Supervivencia , Translocación Genética , Resultado del Tratamiento
4.
Int J Mol Sci ; 20(15)2019 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-31370265

RESUMEN

Osteosarcoma and Ewing sarcoma are the most common malignant primary bone tumors mainly occurring in children, adolescents and young adults. Current standard therapy includes multidrug chemotherapy and/or radiation specifically for Ewing sarcoma, associated with tumor resection. However, patient survival has not evolved for the past decade and remains closely related to the response of tumor cells to chemotherapy, reaching around 75% at 5 years for patients with localized forms of osteosarcoma or Ewing sarcoma but less than 30% in metastatic diseases and patients resistant to initial chemotherapy. Despite Ewing sarcoma being characterized by specific EWSR1-ETS gene fusions resulting in oncogenic transcription factors, currently, no targeted therapy could be implemented. It seems even more difficult to develop a targeted therapeutic strategy in osteosarcoma which is characterized by high complexity and heterogeneity in genomic alterations. Nevertheless, the common point between these different bone tumors is their ability to deregulate bone homeostasis and remodeling and divert them to their benefit. Therefore, targeting different actors of the bone tumor microenvironment has been hypothesized to develop new therapeutic strategies. In this context, it is well known that the Wnt/ß-catenin signaling pathway plays a key role in cancer development, including osteosarcoma and Ewing sarcoma as well as in bone remodeling. Moreover, recent studies highlight the implication of the Wnt/ß-catenin pathway in angiogenesis and immuno-surveillance, two key mechanisms involved in metastatic dissemination. This review focuses on the role played by this signaling pathway in the development of primary bone tumors and the modulation of their specific microenvironment.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Óseas/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/tratamiento farmacológico , Sarcoma de Ewing/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Adolescente , Neoplasias Óseas/genética , Neoplasias Óseas/inmunología , Neoplasias Óseas/mortalidad , Huesos , Niño , Humanos , Metástasis Linfática , Terapia Molecular Dirigida/métodos , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/mortalidad , Neovascularización Patológica/prevención & control , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Osteosarcoma/genética , Osteosarcoma/inmunología , Osteosarcoma/mortalidad , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/inmunología , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Proteína EWS de Unión a ARN/genética , Proteína EWS de Unión a ARN/inmunología , Sarcoma de Ewing/genética , Sarcoma de Ewing/inmunología , Sarcoma de Ewing/mortalidad , Análisis de Supervivencia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Vía de Señalización Wnt/efectos de los fármacos , Adulto Joven , beta Catenina/antagonistas & inhibidores , beta Catenina/genética , beta Catenina/inmunología
5.
BMC Immunol ; 18(1): 40, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28810829

RESUMEN

BACKGROUND: Cancer therapeutic vaccine induced cytotoxic T cell (CTL) responses are pivotal for the killing of tumour cells. Blocking interleukin 10 (IL-10) signalling at the time of immunization increases vaccine induced CTL responses and improves prevention of tumour growth in animal models compared to immunization without an IL-10 signalling blockade. Therefore, this immunization strategy may have potential to curtail cancer in a clinical setting. However, IL-10 deficiency leads to autoimmune disease in the gut. Blocking IL-10 at the time of immunization may result in unwanted side effects, especially immune-pathological diseases in the intestine. METHODS: We investigated whether blocking IL-10 at the time of immunization results in intestinal inflammation responses in a mouse TC-1 tumour model and in a NOD autoimmune disease prone mouse model. RESULTS: We now show that blocking IL-10 at the time of immunization increases IL-10 production by CD4+ T cells in the spleen and draining lymph nodes, and does not result in blood cell infiltration to the intestines leading to intestinal pathological changes. Moreover, immunization with papillomavirus like particles combined with simultaneously blocking IL-10 signalling does not increase the incidence of autoimmune disease in Non-obese diabetic (NOD) mice. CONCLUSIONS: Our results indicate that immunization with an IL-10 inhibitor may facilitate the generation of safe, effective therapeutic vaccines against chronic viral infection and cancer.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Inmunización/efectos adversos , Inmunización/métodos , Interleucina-10/antagonistas & inhibidores , Intestinos/inmunología , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Anticuerpos/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Interleucina-10/inmunología , Interleucina-10/metabolismo , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD/inmunología , Ratones Noqueados , Proteínas de Fusión Oncogénica/inmunología , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/química , Proteínas E7 de Papillomavirus/inmunología , Bazo/citología , Bazo/inmunología
6.
Mol Biol (Mosk) ; 51(2): 274-287, 2017.
Artículo en Ruso | MEDLINE | ID: mdl-28537234

RESUMEN

Early results from clinical trials of autologous chimeric antigen receptor (CAR)-expressing T cells for the therapy of B-cell malignancies have encouraged extending the potency of this therapy to other cancers. However, the success of using CAR T-cells to treat patients with solid tumors has been limited. In this review, we summarize current knowledge on the design and applications of CARs for the targeted therapy of cancer. We describe existing issues that limit the widespread application of CAR T cells and discuss the optimization steps needed to further improve safety and efficacy of this therapeutic platform.


Asunto(s)
Neoplasias Hematológicas , Inmunoterapia/métodos , Proteínas de Fusión Oncogénica , Receptores de Antígenos de Linfocitos T , Linfocitos T , Animales , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/terapia , Humanos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/trasplante
7.
Manag Care ; 26(10): 28-30, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29068298

RESUMEN

FDA approval of the CAR T-cell therapy for leukemia could usher in an era of genetically engineered, individually tailored immunotherapies. But tap those brakes. Long-term results are in short supply-and there's that $475,000 price tag. Or is it a $750,000 price tag?


Asunto(s)
Antineoplásicos/economía , Industria Farmacéutica/economía , Leucemia de Células B/tratamiento farmacológico , Receptores de Antígenos de Linfocitos T , Costos de los Medicamentos , Humanos , Leucemia de Células B/inmunología , Proteínas de Fusión Oncogénica/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/inmunología , Estados Unidos
8.
Blood Cells Mol Dis ; 57: 74-80, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26852660

RESUMEN

With the combination of immunophenotyping and molecular tests, it is still a challenge to identify the characteristics of T cell acute lymphoblastic leukemia (T-ALL) associated with distinct outcomes. This study tests the possible correlation of cellular expression of CD135 and CD117 with somatic gene mutations in T-ALL. One hundred sixty-two samples were tested, including 143 at diagnosis, 15 from T-lymphoblastic lymphoma at relapse, and four relapse samples from sequential follow-up of T-ALL. CD135 and CD117 monoclonal antibodies were included in the T-ALL panel of flow cytometry. The percentage of cells positivity and the median fluorescence intensity were correlated with gene mutational status. STIL-TAL1, TLX3, FLT3 and IL7R mutations were tested using standard techniques. STIL-TAL1 was found in 24.8%, TLX3 in 12%, IL7R in 10% and FLT3-ITD in 5% of cases. FLT3 and IL7R mutations were mutually exclusive, as were FLT3-ITD and STIL-TAL1. Associations of CD135(high) (p<0.01), CD117(intermediate/high) (p=0.02) and FLT3-ITD, CD117(low) with IL7R(mutated) (p<0.01) and CD135(high) with TLX3(pos) were observed. We conclude that the addition of CD135 and CD117 to the diagnosis can predict molecular aberrations in T-ALL settings, mainly segregating patients with FLT3-ITD, who would benefit from treatment with inhibitors of tyrosine.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas de Homeodominio/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteínas Proto-Oncogénicas c-kit/genética , Receptores de Interleucina-7/genética , Tirosina Quinasa 3 Similar a fms/genética , Adolescente , Anticuerpos Monoclonales , Antineoplásicos/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Biomarcadores de Tumor/inmunología , Niño , Preescolar , Femenino , Expresión Génica , Proteínas de Homeodominio/inmunología , Humanos , Inmunofenotipificación , Lactante , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , Masculino , Mutación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-kit/inmunología , Receptores de Interleucina-7/inmunología , Recurrencia , Proteína 1 de la Leucemia Linfocítica T Aguda , Adulto Joven , Tirosina Quinasa 3 Similar a fms/inmunología
9.
Blood ; 123(10): 1535-43, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24449212

RESUMEN

PML-RARA and AML1-ETO are important oncogenic fusion proteins that play a central role in transformation to acute myeloid leukemia (AML). Whether these fusion proteins render the tumor cells with immune evasion properties is unknown. Here we show that both oncogenic proteins specifically downregulate the expression of CD48, a ligand of the natural killer (NK) cell activating receptor 2B4, thereby leading to decreased killing by NK cells. We demonstrate that this process is histone deacetylase (HDAC)-dependent, that it is mediated through the downregulation of CD48 messenger RNA, and that treatment with HDAC inhibitors (HDACi) restores the expression of CD48. Furthermore, by using chromatin immunoprecepitation (ChIP) experiments, we show that AML1-ETO directly interacts with CD48. Finally, we show that AML patients who are carrying these specific translocations have low expression of CD48.


Asunto(s)
Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Escape del Tumor/genética , Escape del Tumor/inmunología , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Secuencia de Bases , Antígeno CD48 , Línea Celular Tumoral , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Citotoxicidad Inmunológica , Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Mieloide Aguda/metabolismo , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1
10.
Adv Exp Med Biol ; 909: 139-67, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27240458

RESUMEN

Cancer is one of the major leading death causes of diseases. Prevention and treatment of cancer is an important way to decrease the incidence of tumorigenesis and prolong patients' lives. Subversive achievements on cancer immunotherapy have recently been paid much attention after many failures in basic and clinical researches. Based on deep analysis of genomics and proteomics of tumor antigens, a variety of cancer vaccines targeting tumor antigens have been tested in preclinical and human clinical trials. Many therapeutic cancer vaccines alone or combination with other conventional treatments for cancer obtained spectacular efficacy, indicating the tremendously potential application in clinic. With the illustration of underlying mechanisms of cancer immune regulation, valid, controllable, and persistent cancer vaccines will play important roles in cancer treatment, survival extension and relapse and cancer prevention. This chapter mainly summarizes the recent progresses and developments on cancer vaccine research and clinical application, thus exploring the existing obstacles in cancer vaccine research and promoting the efficacy of cancer vaccine.


Asunto(s)
Antineoplásicos/uso terapéutico , Vacunas contra el Cáncer/uso terapéutico , Terapia Combinada/métodos , Regulación Neoplásica de la Expresión Génica/inmunología , Factores Inmunológicos/uso terapéutico , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Ratones , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Transducción de Señal , Análisis de Supervivencia
11.
Biochem Biophys Res Commun ; 453(4): 798-803, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25445592

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and its associated receptors (TRAIL-R/TR) are attractive targets for cancer therapy because TRAIL induces apoptosis in tumor cells through TR while having little cytotoxicity on normal cells. Therefore, many agonistic monoclonal antibodies (mAbs) specific for TR have been produced, and these induce apoptosis in multiple tumor cell types. However, some TR-expressing tumor cells are resistant to TR-specific mAb-induced apoptosis. In this study, we constructed a chimeric antigen receptor (CAR) of a TRAIL-receptor 1 (TR1)-specific single chain variable fragment (scFv) antibody (TR1-scFv-CAR) and expressed it on a Jurkat T cell line, the KHYG-1 NK cell line, and human peripheral blood lymphocytes (PBLs). We found that the TR1-scFv-CAR-expressing Jurkat cells killed target cells via TR1-mediated apoptosis, whereas TR1-scFv-CAR-expressing KHYG-1 cells and PBLs killed target cells not only via TR1-mediated apoptosis but also via CAR signal-induced cytolysis, resulting in cytotoxicity on a broader range if target cells than with TR1-scFv-CAR-expressing Jurkat cells. The results suggest that TR1-scFv-CAR could be a new candidate for cancer gene therapy.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Apoptosis/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Proteínas de Fusión Oncogénica/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/inmunología , Proteínas Reguladoras de la Apoptosis/inmunología , Línea Celular Tumoral , Humanos , Células Jurkat
12.
Stem Cells ; 31(2): 236-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23135987

RESUMEN

The initial steps involved in the pathogenesis of acute leukemia are poorly understood. The TEL-AML1 fusion gene usually arises before birth, producing a persistent and covert preleukemic clone that may convert to precursor B cell leukemia following the accumulation of secondary genetic "hits." Here, we show that TEL-AML1 can induce persistent self-renewing pro-B cells in mice. TEL-AML1+ cells nevertheless differentiate terminally in the long term, providing a "window" period that may allow secondary genetic hits to accumulate and lead to leukemia. TEL-AML1-mediated self-renewal is associated with a transcriptional program shared with embryonic stem cells (ESCs), within which Mybl2, Tgif2, Pim2, and Hmgb3 are critical and sufficient components to establish self-renewing pro-B cells. We further show that TEL-AML1 increases the number of leukemia-initiating cells that are generated in collaboration with additional genetic hits, thus providing an overall basis for the development of novel therapeutic and preventive measures targeting the TEL-AML1-associated transcriptional program.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Células Madre Embrionarias/inmunología , Regulación Neoplásica de la Expresión Génica , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Células Precursoras de Linfocitos B/inmunología , Transcripción Genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Células Madre Embrionarias/patología , Feto , Perfilación de la Expresión Génica , Proteína HMGB3/genética , Proteína HMGB3/inmunología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos BALB C , Proteínas de Fusión Oncogénica/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Transducción de Señal , Transactivadores/genética , Transactivadores/inmunología
13.
Biochem J ; 456(3): 361-72, 2013 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-24073693

RESUMEN

Human EMCs (extraskeletal myxoid chondrosarcomas) are soft tissue tumours characterized by specific chromosomal abnormalities. Recently, a proportion of EMCs were found to harbour a characteristic translocation, t(3;9)(q11-12;q22), involving the TFG (TRK-fused gene) at 3q11-12 and the TEC (translocated in extraskeletal chondrosarcoma) gene at 9q22. The present study used both in vitro and in vivo systems to show that the TFG-TEC protein self-associates, and that this is dependent upon the CC (coiled-coil) domain (amino acids 97-124), the AF1 (activation function 1) domain (amino acids 275-562) and the DBD (DNA-binding domain) (amino acids 563-655). The TFG-TEC protein also associated with a mutant NLS-TFG-TEC (AAAA) protein, which harbours mutations in the NLS (nuclear localization signal). Subcellular localization assays showed that the NLS mutant TFG-TEC (AAAA) protein interfered with the nuclear localization of wild-type TFG-TEC. Most importantly, the mutant protein inhibited TFG-TEC-mediated transcriptional activation in vivo. Thus mutations in the TFG-TEC NLS yield a dominant-negative protein. These results show that the biological functions of the TFG-TEC oncogene can be modulated by a dominant-negative mutant.


Asunto(s)
Núcleo Celular , Condrosarcoma , Proteínas de Unión al ADN , Mutación , Neoplasias de los Tejidos Conjuntivo y Blando , Señales de Localización Nuclear , Proteínas de Fusión Oncogénica , Proteínas , Receptores de Esteroides , Receptores de Hormona Tiroidea , Transcripción Genética/genética , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Condrosarcoma/genética , Condrosarcoma/metabolismo , Condrosarcoma/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Neoplasias de los Tejidos Conjuntivo y Blando/genética , Neoplasias de los Tejidos Conjuntivo y Blando/metabolismo , Neoplasias de los Tejidos Conjuntivo y Blando/patología , Señales de Localización Nuclear/genética , Señales de Localización Nuclear/metabolismo , Proteínas de Fusión Oncogénica/inmunología , Proteínas de Fusión Oncogénica/metabolismo , Proteínas/genética , Proteínas/metabolismo , Receptores de Esteroides/genética , Receptores de Esteroides/metabolismo , Receptores de Hormona Tiroidea/genética , Receptores de Hormona Tiroidea/metabolismo
14.
Acta Biochim Biophys Sin (Shanghai) ; 46(1): 6-14, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24240707

RESUMEN

In order to develop more effective therapeutic vaccines against cancers with high-risk human papillomavirus (HPV) infection, it is crucial to enhance the immunogenicity, eliminate the oncogenicity of oncoproteins, and take a combination of E7- and E6-containing vaccines. It has been shown recently that PE(ΔIII)-E7-KDEL3 (E7), a fusion protein containing the HPV16 oncoprotein E7 and the translocation domain of Pseudomonas aeruginosa exotoxin A, is effective against TC-1 tumor cells inoculated in mice, therefore, we engineered PE(ΔIII)-E6-CRL-KDEL3 (E6), the de-oncogenic versions of the E7 and E6 fusion proteins [i.e. PE(ΔIII)-E7(d)-KDEL3, E7(d), and PE(ΔIII)-E6(d)-CRL-KDEL3, E6(d)] and tested the immunoefficacies of these fusion proteins as mono- and bivalent vaccines. Results indicated that the E7(d) get higher immunogenicity than its wild type and the E6 fusion proteins augmented the immunogenicity and antitumor effects of their E7 counterparts. Furthermore, the bivalent vaccine system E7(d) plus E6(d), in the presence of cisplatin, showed the best tumoristatic and tumoricidal effects against established tumors in vivo. Therefore, it can be concluded that this novel therapeutic vaccine system, upon further optimization, may shed new light on clinical management of HPV-related carcinomas.


Asunto(s)
Cisplatino/uso terapéutico , Proteínas de Unión al ADN/uso terapéutico , Proteínas de Fusión Oncogénica/uso terapéutico , Proteínas Oncogénicas Virales/uso terapéutico , Proteínas E7 de Papillomavirus/uso terapéutico , Vacunas contra Papillomavirus/uso terapéutico , ADP Ribosa Transferasas/uso terapéutico , Animales , Antineoplásicos/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Sinergismo Farmacológico , Exotoxinas/uso terapéutico , Ratones , Proteínas de Fusión Oncogénica/inmunología , Vacunas contra Papillomavirus/inmunología , Factores de Virulencia/uso terapéutico , Exotoxina A de Pseudomonas aeruginosa
15.
Biol Blood Marrow Transplant ; 19(6): 958-66, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23542687

RESUMEN

Using real-time quantitative PCR, we monitored Wilms tumor gene 1 (WT1) expression from diagnosis to hematopoietic stem cell transplantation (HSCT) in adult patients with cytogenetically normal acute myelogenous leukemia (CN-AML) and FLT3-ITD and NPM1 mutations. The values at diagnosis were evaluated in 104 patients. Data collected after induction chemotherapy were available for all patients, but only 68 patients were treated with HSCT. Significant WT1 expression cut-offs were determined by receiver operation characteristic curve analysis, and rates of overall survival (OS) and disease-free survival (DFS) were estimated. WT1 decrement ratios (DR) at postinduction chemotherapy and at pre- and post-HSCT compared with the diagnostic level were calculated. Higher WT1 expression at diagnosis, postinduction chemotherapy, and pre-HSCT showed inferior OS (P = .015, <.001, and .002) and DFS (P = .006, <.001, and .003). The cut-offs were determined at the median for diagnostic WT1 expression and at the 25% level from the top for other time points excluding post-HSCT. The WT1 DR ≥ 1-log after induction chemotherapy showed superior OS and DFS (P = .009 and .002) and WT1 DR ≥ 1-log preceding HSCT also showed superior OS and DFS (P = .009 and .003). Results of WT1 DR were consistently applicable in each subgroup with higher (≥ 1.0) and lower (<1.0) WT1 expression at diagnosis and also in NPM1-wild-type/FLT3-ITD-negative CN-AML. The WT1 DR therefore predicted survival outcomes after HSCT more accurately than did the diagnostic WT1 expression. WT1 expression may serve as a reliable marker for residual disease and WT1 DR as a prognostic indicator, particularly in NPM1-wild-type/FLT3-ITD-negative CN-AML. These measures may be applied throughout the course of treatment and even after HSCT.


Asunto(s)
Biomarcadores de Tumor/inmunología , Enfermedad Injerto contra Huésped/diagnóstico , Trasplante de Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/diagnóstico , Neoplasia Residual/diagnóstico , Acondicionamiento Pretrasplante , Proteínas WT1/inmunología , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/genética , Análisis Citogenético , Femenino , Expresión Génica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/terapia , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Agonistas Mieloablativos/uso terapéutico , Neoplasia Residual/inmunología , Neoplasia Residual/mortalidad , Neoplasia Residual/terapia , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Nucleofosmina , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Recurrencia , Estudios Retrospectivos , Análisis de Supervivencia , Trasplante Homólogo , Resultado del Tratamiento , Proteínas WT1/genética
16.
Cancer Immunol Immunother ; 62(12): 1831-40, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24149465

RESUMEN

Identification of novel vaccine targets is critical for the design and advancement of prostate cancer (PCa) immunotherapy. Ideal targets are proteins that are abundant in prostate tumors while absent in extra-prostatic tissues. The fusion of the androgen-regulated TMPRSS2 gene with the ETS transcription factor ERG occurs in approximately 50 % of prostate cancer cases and results in aberrant ERG expression. Because expression of ERG is very low in peripheral tissue, we evaluated the suitability of this protein as an antigen target in PCa vaccines. ERG-derived HLA-A*0201-restricted immunogenic epitopes were identified through a 3-step strategy that included in silico, in vitro, and in vivo validation. Algorithms were used to predict potential HLA-A*0201-binding epitopes. High-scoring epitopes were tested for binding to HLA-A*0201 using the T2-based stabilization assay in vitro. Five peptides were found to bind HLA-A*0201 and were subsequently tested for immunogenicity in humanized, HLA-A*0201 transgenic mice. The in vivo screening identified three immunogenic peptides. One of these peptides, ERG295, overcame peripheral tolerance in HLA-A*0201 mice that expressed prostate-restricted ERG. Also, this peptide induced an antigen-specific response against ERG-expressing human prostate tumor cells. Finally, tetramer assay showed detectable and responsive ERG295-specific cytotoxic lymphocytes in peripheral blood of HLA-A*0201(+) prostate cancer patients. Detection of ERG-specific CTLs in both mice and the blood of prostate cancer patients indicates that ERG-specific tolerance can be overcome. Additionally, these data suggest that ERG is a suitable target antigen for PCa immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Antígeno HLA-A2/inmunología , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Neoplasias de la Próstata/terapia , Linfocitos T Citotóxicos/inmunología , Animales , Estudios de Casos y Controles , Epítopos de Linfocito T/inmunología , Citometría de Flujo , Humanos , Inmunoterapia , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/metabolismo
17.
Blood ; 118(4): 946-54, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21613253

RESUMEN

Adoptive therapy with T-cell receptor (TCR)-engineered T cells is a promising approach in cancer treatment. While usage of T cells specific for tumor-associated antigens (TAAs) can lead to serious side effects because of autoimmunity, targeting true tumor-specific mutations, such as the products of translocations in leukemias, should reduce such a risk. A potentially ideal target might be the chimeric protein TEL-AML1, which results from the chromosomal translocation 12;21 and represents the most common fusion gene in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Within the fusion region of TEL-AML1, a single epitope has been described by reverse immunology as immunogenic in HLA-A*0201 restriction settings. As a potential source of TCRs specific for this TEL-AML1 epitope, we have used mice expressing a human TCR-αß repertoire and human MHC class I. Surprisingly, we have found that, although a specific functional CD8(+) T-cell response against this peptide could be evoked, the described epitope was in fact not endogenously processed. Analyses done with a potent antigen-presenting cell line, as well as with purified human proteasomes, support the conclusion that this peptide cannot be proposed as a potential target in immunotherapy of ALL in HLA-A*0201-restricted fashion.


Asunto(s)
Presentación de Antígeno/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Epítopos de Linfocito T/inmunología , Proteínas de Fusión Oncogénica/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Separación Celular , Cromatografía Líquida de Alta Presión , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Translocación Genética
18.
Mol Cell Probes ; 27(1): 1-5, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22906630

RESUMEN

Using an acute promyelocytic leukemia (APL) preclinical model, we show that oncogene-specific PCR (Polymerase Chain Reaction)-based assays allow to evaluate the efficacy of immunotherapy combining all-trans retinoic acid (ATRA) and a DNA-based vaccine targeting the promyelocytic leukemia-retinoic acid receptor alpha (PML-RARα) oncogene. Kaplan-Meier survival analysis according to the peripheral blood PML-RARα normalized copy number (NCN) clearly shows that ATRA + DNA-treated mice with an NCN lower than 10 (43%) formed the group with a highly significant (p < 0.0001) survival advantage. Furthermore, a PCR assay was used to assess various tissues and organs for the presence of PML-RARα-positive cells in long-term survivors (n = 15). As expected, the majority of mice (n = 10) had no measurable tissue level of PML-RARα. However, five mice showed a weak positive signal in both the brain and spleen (n = 2), in the brain only (n = 2) and in the spleen only (n = 1). Thus tracking the oncogene-positive cells in long-term survivors reveals for the first time that extramedullary PML-RARα-positive cell reservoirs such as the brain may persist and be involved in relapses.


Asunto(s)
Inmunoterapia , Leucemia Promielocítica Aguda/terapia , Proteínas de Fusión Oncogénica/metabolismo , Tretinoina/uso terapéutico , Vacunas de ADN/uso terapéutico , Animales , Encéfalo/citología , Dosificación de Gen , Estimación de Kaplan-Meier , Leucemia Promielocítica Aguda/mortalidad , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/uso terapéutico , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Bazo/citología , Resultado del Tratamiento
19.
J Immunol ; 187(4): 1653-63, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21768400

RESUMEN

Graft-versus-leukemia (GVL) against chronic-phase chronic myelogenous leukemia (CP-CML) is potent, but it is less efficacious against acute leukemias and blast-crisis chronic myelogenous leukemia (BC-CML). The mechanisms underlying GVL resistance are unknown. Previously, we found that alloreactive T cell targeting of GVL-sensitive bcr-abl-induced mouse CP-CML (mCP-CML) required TCR-MHC interactions and that multiple and redundant killing mechanisms were in play. To better understand why BC-CML is resistant to GVL, we performed a comprehensive analysis of GVL against mouse BC-CML (mBC-CML) induced by the retroviral transfer of the bcr-abl and NUP98/HOXA9 fusion cDNAs. Like human BC-CML, mBC-CML was GVL resistant, and this was not due to accelerated kinetics or a greater leukemia burden. To study T cell recognition and killing mechanisms, we generated a panel of gene-deficient leukemias by transducing bone marrow from gene-deficient mice. T cell target recognition absolutely required that mBC-CML cells express MHC molecules. GVL against both mCP-CML and mBC-CML required leukemia expression of ICAM-1. We hypothesized that mBC-CML would be resistant to some of the killing mechanisms sufficient to eliminate mCP-CML, but we found instead that the same mechanisms were effective against both types of leukemia, because GVL was similar against wild-type or mBC-CML genetically lacking Fas, TRAIL-R, Fas/TRAIL-R, or TNFR1/R2 or when donor T cells were perforin(-/-). However, mCP-CML, but not mBC-CML, relied on expression of programmed death-1 ligands 1 and 2 (PD-L1/L2) to resist T cell killing, because only GVL against mCP-CML was augmented when leukemias lacked PD-L1/L2. Thus, mBC-CML cells have cell-intrinsic mechanisms, distinct from mCP-CML cells, which protect them from T cell killing.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/inmunología , Crisis Blástica/inmunología , Efecto Injerto vs Leucemia/inmunología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Crisis Blástica/genética , Genes abl/inmunología , Efecto Injerto vs Leucemia/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/inmunología , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Retroviridae , Linfocitos T/inmunología , Transducción Genética
20.
Cancer Sci ; 103(9): 1625-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22726592

RESUMEN

In the present study, we evaluated the safety and effectiveness of SYT-SSX-derived peptide vaccines in patients with advanced synovial sarcoma. A 9-mer peptide spanning the SYT-SSX fusion region (B peptide) and its HLA-A*2402 anchor substitute (K9I) were synthesized. In Protocols A1 and A2, vaccines with peptide alone were administered subcutaneously six times at 14-day intervals. The B peptide was used in Protocol A1, whereas the K9I peptide was used in Protocol A2. In Protocols B1 and B2, the peptide was mixed with incomplete Freund's adjuvant and then administered subcutaneously six times at 14-day intervals. In addition, interferon-α was injected subcutaneously on the same day and again 3 days after the vaccination. The B peptide and K9I peptide were used in Protocols B1 and B2, respectively. In total, 21 patients (12 men, nine women; mean age 43.6 years) were enrolled in the present study. Each patient had multiple metastatic lesions of the lung. Thirteen patients completed the six-injection vaccination schedule. One patient developed intracerebral hemorrhage after the second vaccination. Delayed-type hypersensitivity skin tests were negative in all patients. Nine patients showed a greater than twofold increase in the frequency of CTLs in tetramer analysis. Recognized disease progression occurred in all but one of the nine patients in Protocols A1 and A2. In contrast, half the 12 patients had stable disease during the vaccination period in Protocols B1 and B2. Of note, one patient showed transient shrinkage of a metastatic lesion. The response of the patients to the B protocols is encouraging and warrants further investigation.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Proteínas de Fusión Oncogénica/inmunología , Sarcoma Sinovial/tratamiento farmacológico , Vacunas de Subunidad/uso terapéutico , Adulto , Anciano , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , Femenino , Antígenos HLA-A/inmunología , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Masculino , Persona de Mediana Edad , Sarcoma Sinovial/inmunología , Sarcoma Sinovial/patología , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/efectos adversos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA