Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Acta Med Okayama ; 74(5): 371-379, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33106692

RESUMEN

The prognosis of advanced non-small cell lung cancer (NSCLC) patients has improved in recent decades, especially for patients with an oncogenic driver mutation. Anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors (TKIs) are effective for patients with the echinoderm microtubule-associated protein-like 4-ALK fusion gene. Several ALK-TKIs have been established: the first-generation ALK-TKI, crizotinib; second-generation ALK-TKIs, alectinib and ceritinib; and third-generation ALK-TKI, lorlatinib. Some ALK-TKIs are effective for tumors that are resistant to other ALK-TKIs; however, as is known in epidermal growth factor receptormutant lung cancer, tumor resistance is inevitable. ALK-positive NSCLCs acquire resistance via various mechanisms, making it a heterogeneous disease. Therefore, it is necessary to develop next-generation treatment strategies, such as the use of next-generation ALK-TKIs for secondary mutations, or combination therapies with ALK-TKIs and other TKIs. In this review, we summarize the development and use of ALK-TKIs, prior pivotal clinical trials, and resistance mechanisms.


Asunto(s)
Quinasa de Linfoma Anaplásico/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/administración & dosificación , Quinasa de Linfoma Anaplásico/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Inhibidores de Proteínas Quinasas/metabolismo
2.
Invest New Drugs ; 38(2): 457-467, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31065954

RESUMEN

Background Fibroblast growth factors (FGFs) have a fundamental role in cancer. Sequestering FGFs with GSK3052230 (FP-1039) blocks their ability to activate FGFRs while avoiding toxicities associated with small molecule inhibitors of FGFR, including hyperphosphatemia and retinal, nail, and skin toxicities. Methods A multicenter, open-label, phase Ib study evaluated weekly GSK3052230 added to pemetrexed/cisplatin in patients with treatment-naive, unresectable malignant pleural mesothelioma. Doses were escalated according to a 3 + 3 design, followed by cohort expansion at the maximum tolerated dose (MTD). Endpoints included safety, overall response rate, progression-free survival, and pharmacokinetics. Results 36 patients were dosed at 10, 15, and 20 mg/kg doses of GSK3052230. Three dose-limiting toxicities were observed at 20 mg/kg and one at 15 mg/kg. The MTD was defined as 15 mg/kg and used for cohort expansion. The most common treatment-related adverse events (AEs) were nausea (56%), decreased appetite (36%), infusion reactions (36%), decreased neutrophil counts (36%), and fatigue (33%). The confirmed ORR was 39% (95% CI: 23.1-56.5) (14/36 PRs) and 47% had stable disease (17/36), giving a disease control rate of 86%. At 15 mg/kg GSK3052230 (n = 25), the ORR was 44% (95% CI: 24.4-65.1), and the median PFS was 7.4 months (95% CI: 6.7-13.4). Four patients had disease control for over 1 year, and three were still ongoing. Conclusion At 15 mg/kg weekly, GSK3052230 was well tolerated in combination with pemetrexed/cisplatin and durable responses were observed. Importantly, AEs associated with small molecule inhibitors of FGFR were not observed, as predicted by the unique mechanism of action of this drug.


Asunto(s)
Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cisplatino/administración & dosificación , Inmunoglobulina G/administración & dosificación , Mesotelioma Maligno/tratamiento farmacológico , Proteínas de Fusión Oncogénica/administración & dosificación , Pemetrexed/administración & dosificación , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Cisplatino/efectos adversos , Femenino , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Inmunoglobulina G/efectos adversos , Ligandos , Masculino , Mesotelioma Maligno/metabolismo , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas de Fusión Oncogénica/farmacocinética , Pemetrexed/efectos adversos , Proteínas Recombinantes de Fusión , Resultado del Tratamiento
3.
Int J Oncol ; 51(3): 949-958, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28677808

RESUMEN

Breast cancer is the most common carcinoma among Chinese women. Interferon α (IFNα) has been used to treat various types of cancer, including breast cancer, but its antitumor activity is relative low, which significantly hinders its clinical application. In this study, we utilized a Ph.D.-12 peptide library screening system to identify a short peptide that specifically binds to MCF-7 breast cancer cells. By fusing the MCF-7 binding peptide (MBP) to the C-terminus of IFNα, we constructed an engineered IFNα-MBP fusion molecule (IMBP), and applied this novel fusion protein to the treatment of breast cancer. We found that IMBP exhibited significantly higher activity than wild-type IFNα in inhibiting cell growth and inducing cell apoptosis. Additionally, IMBP potentiated the therapeutic efficacy of doxorubicin-based breast cancer chemotherapy via the activation of cell cycle arrest and cell apoptosis pathway genes including p53, p21, CDK2, cyclin A, caspase 9, Bcl-2 and Bax. The enhanced activity of the synthetic IMBP was also associated with the activation of signal transducer and activation of transcription 1 (STAT1) pathway target genes (STAT1, IFIT1, IFITM1 and MX1). This study evaluated the potential value of the synthetic IMBP as a novel anti-breast cancer agent.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Doxorrubicina/administración & dosificación , Interferón-alfa/administración & dosificación , Proteínas de Neoplasias/genética , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Doxorrubicina/efectos adversos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Interferón-alfa/química , Interferón-alfa/genética , Células MCF-7 , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/química , Proteínas de Fusión Oncogénica/genética , Biblioteca de Péptidos , Unión Proteica
4.
Clin Cancer Res ; 23(14): 3499-3509, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28183713

RESUMEN

Purpose: The multiple mechanisms used by solid tumors to suppress tumor-specific immune responses are a major barrier to the success of adoptively transferred tumor-specific T cells. As viruses induce potent innate and adaptive immune responses, we hypothesized that the immunogenicity of viruses could be harnessed for the treatment of solid tumors if virus-specific T cells (VST) were modified with tumor-specific chimeric antigen receptors (CAR). We tested this hypothesis using VZV-specific T cells (VZVST) expressing a CAR for GD2, a disialoganglioside expressed on neuroblastoma and certain other tumors, so that the live-attenuated VZV vaccine could be used for in vivo stimulation.Experimental Design: We generated GMP-compliant, GD2.CAR-modified VZVSTs from healthy donors and cancer patients by stimulation of peripheral blood mononuclear cells with overlapping peptide libraries spanning selected VZV antigens, then tested their ability to recognize and kill GD2- and VZV antigen-expressing target cells.Results: Our choice of VZV antigens was validated by the observation that T cells specific for these antigens expanded in vivo after VZV vaccination. VZVSTs secreted cytokines in response to VZV antigens, killed VZV-infected target cells and limited infectious virus spread in autologous fibroblasts. However, while GD2.CAR-modified VZVSTs killed neuroblastoma cell lines on their first encounter, they failed to control tumor cells in subsequent cocultures. Despite this CAR-specific dysfunction, CAR-VZVSTs retained functional specificity for VZV antigens via their TCRs and GD2.CAR function was partially rescued by stimulation through the TCR or exposure to dendritic cell supernatants.Conclusions: Vaccination via the TCR may provide a means to reactivate CAR-T cells rendered dysfunctional by the tumor microenvironment (NCT01953900). Clin Cancer Res; 23(14); 3499-509. ©2017 AACR.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Proteínas de Fusión Oncogénica/administración & dosificación , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Adolescente , Adulto , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/inmunología , Femenino , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Masculino , Ratones , Persona de Mediana Edad , Neuroblastoma/inmunología , Neuroblastoma/patología , Proteínas de Fusión Oncogénica/inmunología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Microambiente Tumoral/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Clin Cancer Res ; 23(13): 3416-3427, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28069723

RESUMEN

Purpose: To generate and characterize a murine GITR ligand fusion protein (mGITRL-FP) designed to maximize valency and the potential to agonize the GITR receptor for cancer immunotherapy.Experimental Design: The EC50 value of the mGITRL-FP was compared with an anti-GITR antibody in an in vitro agonistic cell-based reporter assay. We assessed the impact of dose, schedule, and Fc isotype on antitumor activity and T-cell modulation in the CT26 tumor model. The activity of the mGITRL-FP was compared with an agonistic murine OX40L-FP targeting OX40, in CT26 and B16F10-Luc2 tumor models. Combination of the mGITRL-FP with antibodies targeting PD-L1, PD-1, or CTLA-4 was analyzed in mice bearing CT26 tumors.Results: The mGITRL-FP had an almost 50-fold higher EC50 value compared with an anti-murine GITR antibody. Treatment of CT26 tumor-bearing mice with mGITRL-FP-mediated significant antitumor activity that was dependent on isotype, dose, and duration of exposure. The antitumor activity could be correlated with the increased proliferation of peripheral CD8+ and CD4+ T cells and a significant decrease in the frequency of intratumoral Tregs. The combination of mGITRL-FP with mOX40L-FP or checkpoint inhibitor antagonists enhanced antitumor immunity above that of monotherapy treatment.Conclusions: These results suggest that therapeutically targeting GITR represents a unique approach to cancer immunotherapy and suggests that a multimeric fusion protein may provide increased agonistic potential versus an antibody. In addition, these data provide, for the first time, early proof of concept for the potential combination of GITR targeting agents with OX40 agonists and PD-L1 antagonists. Clin Cancer Res; 23(13); 3416-27. ©2017 AACR.


Asunto(s)
Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Melanoma Experimental/inmunología , Proteínas de Fusión Oncogénica/administración & dosificación , Factores de Necrosis Tumoral/inmunología , Animales , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Modelos Animales de Enfermedad , Proteína Relacionada con TNFR Inducida por Glucocorticoide/administración & dosificación , Humanos , Melanoma Experimental/genética , Melanoma Experimental/terapia , Glicoproteínas de Membrana/agonistas , Glicoproteínas de Membrana/inmunología , Ratones , Ligando OX40 , Proteínas de Fusión Oncogénica/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Factores de Necrosis Tumoral/agonistas , Factores de Necrosis Tumoral/genética
6.
Mol Ther ; 24(8): 1412-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27166877

RESUMEN

The EWS/FLI1 fusion gene is well characterized as a driver of Ewing's sarcoma. Bi-shRNA EWS/FLI1 is a functional plasmid DNA construct that transcribes both siRNA and miRNA-like effectors each of which targets the identical type 1 translocation junction region of the EWS/FLI1 transcribed mRNA sequence. Previous preclinical and clinical studies confirm the safety of this RNA interference platform technology and consistently demonstrate designated mRNA and protein target knockdown at greater than 90% efficiency. We initiated development of pbi-shRNA EWS/FLI1 lipoplex (LPX) for the treatment of type 1 Ewing's sarcoma. Clinical-grade plasmid was manufactured and both sequence and activity verified. Target protein and RNA knockdown of 85-92% was demonstrated in vitro in type 1 human Ewing's sarcoma tumor cell lines with the optimal bi-shRNA EWS/FLI1 plasmid. This functional plasmid was placed in a clinically tested, liposomal (LP) delivery vehicle followed by in vivo verification of activity. Type 1 Ewing's sarcoma xenograft modeling confirmed dose related safety and tumor response to pbi-shRNA EWS/FLI1 LPX. Toxicology studies in mini-pigs with doses comparable to the demonstrated in vivo efficacy dose resulted in transient fever, occasional limited hypertension at low- and high-dose assessment and transient liver enzyme elevation at high dose. These results provide the justification to initiate clinical testing.


Asunto(s)
Liposomas , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , ARN Interferente Pequeño/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/genética , Animales , Sitios de Unión , Línea Celular Tumoral , Citocinas/metabolismo , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Técnicas de Silenciamiento del Gen , Orden Génico , Terapia Genética , Humanos , Mediadores de Inflamación , Masculino , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/química , Plásmidos/administración & dosificación , Plásmidos/genética , Proteína Proto-Oncogénica c-fli-1/administración & dosificación , Proteína Proto-Oncogénica c-fli-1/química , Interferencia de ARN , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/química , Proteína EWS de Unión a ARN/administración & dosificación , Proteína EWS de Unión a ARN/química , Sarcoma de Ewing/mortalidad , Sarcoma de Ewing/patología , Sarcoma de Ewing/terapia , Reparación del Gen Blanco , Transfección , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
ACS Appl Mater Interfaces ; 8(14): 8918-27, 2016 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-26890991

RESUMEN

Tumor stromal cells have been increasingly recognized to interact with tumor parenchyma cells and promote tumor growth. Therefore, we speculated that therapeutics delivery to both parenchyma cells and stromal cells simultaneously might treat a tumor more effectively. Tissue factor (TF) was shown to be extensively located in a tumor and was abundantly sited in both tumor parenchyma cells and stromal cells including neo-vascular cells, tumor-associated fibroblasts, and tumor-associated macrophages, indicating it might function as a favorable target for drug delivery to multiple cell types simultaneously. EGFP-EGF1 is a fusion protein derived from factor VII, the natural ligand of TF. It retains the specific TF binding capability but does not cause coagulation. In the present study, a nanoparticle modified with EGFP-EGF1 (ENP) was constructed as a multitargeting drug delivery system. The protein binding experiment showed EGFP-EGF1 could bind well to A549 tumor cells and other stromal cells including neo-vascular cells, tumor-associated fibroblasts, and tumor-associated macrophages. Compared with unmodified nanoparticles (NP), ENP uptake by A549 cells and those stromal cells was significantly enhanced but inhibited by excessive free EGFP-EGF1. In addition, ENP induced more A549 tumor cell apoptosis than Taxol and NP when paclitaxel (PTX) was loaded. In vivo, ENP accumulated more specially in TF-overexpressed A549 tumors by in vivo imaging, mainly regions unoccupied by factor VII and targeted tumor parenchyma cells as well as different types of stromal cells by immunofluorescence staining. Treatment with PTX-loaded ENP (ENP-PTX) significantly reduced the A549 tumor growth in nude mice while NP-PTX- and Taxol-treated mice had lower response to the therapy. Furthermore, H&E and TUNEL staining revealed that ENP-PTX induced more severe tumor necrosis and more extensive cell apoptosis. Altogether, the present study demonstrated that ENP could target multiple key cell types in tumors through TF, which could be utilized to improve the therapeutic effect of anticancer drugs.


Asunto(s)
Sistemas de Liberación de Medicamentos , Proteínas Fluorescentes Verdes/genética , Proteínas Mitocondriales/genética , Nanopartículas/administración & dosificación , Neoplasias/tratamiento farmacológico , Factor G de Elongación Peptídica/genética , Células A549 , Animales , Apoptosis/efectos de los fármacos , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Proteínas Fluorescentes Verdes/administración & dosificación , Proteínas Fluorescentes Verdes/química , Humanos , Macrófagos/efectos de los fármacos , Ratones , Proteínas Mitocondriales/administración & dosificación , Proteínas Mitocondriales/química , Nanopartículas/química , Neoplasias/patología , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/química , Paclitaxel/administración & dosificación , Paclitaxel/química , Factor G de Elongación Peptídica/administración & dosificación , Factor G de Elongación Peptídica/química , Unión Proteica , Células del Estroma/efectos de los fármacos , Células del Estroma/patología
8.
Oncotarget ; 6(35): 37678-94, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26462019

RESUMEN

Ewing sarcoma is an aggressive tumor of bone and soft tissue affecting predominantly children and young adults. Tumor-specific chromosomal translocations create EWS-FLI1 and similar aberrant ETS fusion proteins that drive sarcoma development in patients. ETS family fusion proteins and over-expressed ETS proteins are also found in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) patients. Transgenic expression of EWS-FLI1 in mice promotes high penetrance erythroid leukemia with dense hepatic and splenic infiltrations. We identified a small molecule, YK-4-279, that directly binds to EWS-FLI1 and inhibits its oncogenic activity in Ewing sarcoma cell lines and xenograft mouse models. Herein, we tested in vivo therapeutic efficacy and potential side effects of YK-4-279 in the transgenic mouse model with EWS-FLI1 induced leukemia. A two-week course of treatment with YK-4-279 significantly reduced white blood cell count, nucleated erythroblasts in the peripheral blood, splenomegaly, and hepatomegaly of erythroleukemic mice. YK-4-279 inhibited EWS-FLI1 target gene expression in neoplastic cells. Treated animals showed significantly better overall survival compared to control mice that rapidly succumbed to leukemia. YK-4-279 treated mice did not show overt toxicity in liver, spleen, or bone marrow. In conclusion, this in vivo study highlights the efficacy of YK-4-279 to treat EWS-FLI1 expressing neoplasms and support its therapeutic potential for patients with Ewing sarcoma and other ETS-driven malignancies.


Asunto(s)
Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indoles/farmacología , Leucemia Eritroblástica Aguda/tratamiento farmacológico , Leucemia Eritroblástica Aguda/etiología , Proteínas de Fusión Oncogénica/antagonistas & inhibidores , Proteínas de Fusión Oncogénica/toxicidad , Proteína Proto-Oncogénica c-fli-1/antagonistas & inhibidores , Proteína Proto-Oncogénica c-fli-1/toxicidad , Proteína EWS de Unión a ARN/antagonistas & inhibidores , Proteína EWS de Unión a ARN/toxicidad , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Leucemia Eritroblástica Aguda/patología , Ratones , Ratones Transgénicos , Proteínas de Fusión Oncogénica/administración & dosificación , Proteína Proto-Oncogénica c-fli-1/administración & dosificación , ARN Mensajero/genética , Proteína EWS de Unión a ARN/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Resonancia por Plasmón de Superficie
9.
Brain Res ; 1390: 59-69, 2011 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-21443868

RESUMEN

The development of the cancer stem cell (CSCs) niche theory has provided a new target for the treatment of gliomas. Gene therapy using oncolytic viral vectors has shown great potential for the therapeutic targeting of CSCs. To explore whether a viral vector carrying an exogenous Endo-Angio fusion gene (VAE) can infect and kill glioma stem cells (GSCs), as well as inhibit their vascular niche in vitro, we have collected surgical specimens of human high-grade glioma (world health organization, WHO Classes III-VI) from which we isolated and cultured GSCs under conditions originally designed for the selective expansion of neural stem cells. Our results demonstrate the following: (1) Four lines of GSCs (isolated from 20 surgical specimens) could grow in suspension, were multipotent, had the ability to self-renew and expressed the neural stem cell markers, CD133 and nestin. (2) VAE could infect GSCs and significantly inhibit their viability. (3) The Endo-Angio fusion gene was expressed in GSCs 48 h after VAE infection and could inhibit the proliferation of human brain microvascular endothelial cells (HBMEC). (4) Residual viable cells lose the ability of self-renewal and adherent differentiation. In conclusion, VAE can significantly inhibit the activity of GSCs in vitro and the expression of exogenous Endo-Angio fusion gene can inhibit HBMEC proliferation. VAE can be used as a novel virus-gene therapy strategy for glioma.


Asunto(s)
Angiostatinas/genética , Endostatinas/genética , Glioma/genética , Células Madre Neoplásicas , Virus Oncolíticos/genética , Angiostatinas/administración & dosificación , Animales , Supervivencia Celular/genética , Células Cultivadas , Endostatinas/administración & dosificación , Terapia Genética/métodos , Terapia Genética/tendencias , Glioma/patología , Glioma/terapia , Humanos , Células Madre Neoplásicas/patología , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Ratas , Células Tumorales Cultivadas
10.
Blood ; 115(3): 653-6, 2010 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-19965687

RESUMEN

DNA vaccination and all-trans retinoic acid (ATRA) result in a survival advantage in a mouse model of acute promyelocytic leukemia (APL). Depletion of CD4(+) or CD8(+) cells abolished this effect. CD4(+) depletions of long-term survivors resulted in relapse and death within 3 months, thus demonstrating the need of both CD4(+) and CD8(+) subsets for the generation of DNA-driven antileukemic immune responses and underscoring a crucial role of CD4(+) cells in the maintenance of durable remissions. Degranulation and cytotoxic carboxyfluorescein diacetate succinimidyl ester-based assays showed major histocompatibility complex-restricted APL-specific T cell-mediated immune responses. Sorted APL-specific CD8(+)CD107a(+) T cells showed an increase of antileukemic activity. Effectors from ATRA + DNA-treated mice were shown to secrete interferon-gamma when stimulated with either APL cells or peptides from the promyelocytic leukemia-RARalpha vaccine-derived sequences as detected by ELISpot assays. Our results demonstrate that DNA vaccination with ATRA confers the effective boosting of interferon-gamma-producing and cytotoxic T cells in the leukemic mice.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Leucemia Promielocítica Aguda/terapia , Tretinoina/administración & dosificación , Vacunas de ADN/administración & dosificación , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Terapia Combinada , Modelos Animales de Enfermedad , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Leucemia Promielocítica Aguda/inmunología , Leucemia Promielocítica Aguda/patología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/genética , Ratones , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Análisis de Supervivencia , Resultado del Tratamiento , Células Tumorales Cultivadas , Vacunas de ADN/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Mol Cancer Ther ; 7(5): 1329-35, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18483320

RESUMEN

Genital human papillomavirus (HPV) infection is the primary cause of cervical cancer in women. Although the HPV recombinant L1 protein was recently licensed as an available vaccine, it has numerous shortcomings. New vaccination strategies should be considered. To enable the design of a prophylactic and therapeutic low-cost vaccine candidate, chimeric HPV16 L1DeltaC34E7N1-60 capsomeres were produced in Escherichia coli. The immune characteristics and potential prophylactic and therapeutic effects of these capsomeres were examined in C57BL/6 mice. Following protein purification and renaturation, the majority of the recombinant chimeric proteins (L1DeltaC34E7N1-60) assembled into capsomeres. These capsomeres were able to induce conformational and neutralizing antibodies against HPV virus-like particles and trigger cell-mediated specific immune responses against the L1 and E7 peptides. In vivo tumor challenge assays showed that mice immunized with the capsomeres were protected against a challenge with both C3 and TC-1 tumor cells. Furthermore, in vivo tumor rejection assays showed that capsomeres have therapeutic efficacy in mice following inoculation with C3 and TC-1 tumor cells. Chimeric capsomeres are capable of preventing and eliminating HPV16 infection. Therefore, our study has provided an economical vaccine candidate.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas de Fusión Oncogénica/uso terapéutico , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/uso terapéutico , Infecciones por Papillomavirus/tratamiento farmacológico , Vacunas contra Papillomavirus/uso terapéutico , Animales , Proteínas de la Cápside/metabolismo , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Proteínas Oncogénicas Virales/administración & dosificación , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Infecciones Tumorales por Virus/tratamiento farmacológico , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/prevención & control
12.
Int J Cancer ; 121(12): 2794-800, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17721997

RESUMEN

Persistent infection with human papillomaviruses (HPV) is a prerequisite for the development of cervical cancer. Vaccination with virus-like particles (VLP) has demonstrated efficacy in prophylaxis but lacks therapeutic potential. HPV16 L1E7 chimeric virus-like particles (CVLP) consist of a carboxy-terminally truncated HPV16L1 protein fused to the amino-terminal part of the HPV16 E7 protein and self-assemble by recombinant expression of the fusion protein. The CVLP are able to induce L1- and E7-specific cytotoxic T lymphocytes. We have performed a first clinical trial to gain information about the safety and to generate preliminary data on the therapeutic potential of the CVLP in humans. A randomized, double blind, placebo-controlled clinical trial has been conducted in 39 HPV16 mono-infected high grade cervical intraepithelial neoplasia (CIN) patients (CIN 2/3). Two doses (75 mug or 250 mug) of CVLP were applied. The duration of the study was 24 weeks with 2 optional visits after another 12 and 24 weeks. The vaccine showed a very good safety profile with only minor adverse events attributable to the immunization. Antibodies with high titers against HPV16 L1 and low titers against HPV16 E7 as well as cellular immune responses against both proteins were induced. Responses were equivalent for both vaccine concentrations. A trend for histological improvement to CIN 1 or normal was seen in 39% of the patients receiving the vaccine and only 25% of the placebo recipients. Fifty-six percent of the responders were also HPV16 DNA-negative by the end of the study. Therefore, we demonstrated evidence for safety and a nonsignificant trend for the clinical efficacy of the HPV16 L1E7 CVLP vaccine.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Papillomavirus Humano 16/inmunología , Proteínas de Fusión Oncogénica/uso terapéutico , Proteínas Oncogénicas Virales/uso terapéutico , Vacunas contra Papillomavirus/uso terapéutico , Displasia del Cuello del Útero/tratamiento farmacológico , Displasia del Cuello del Útero/virología , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/virología , Adulto , Anciano , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/efectos adversos , ADN Viral/efectos de los fármacos , ADN Viral/aislamiento & purificación , Método Doble Ciego , Esquema de Medicación , Femenino , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/aislamiento & purificación , Humanos , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/efectos adversos , Proteínas Oncogénicas Virales/administración & dosificación , Proteínas Oncogénicas Virales/efectos adversos , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/tratamiento farmacológico , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/tratamiento farmacológico , Infecciones Tumorales por Virus/inmunología , Neoplasias del Cuello Uterino/inmunología , Neoplasias del Cuello Uterino/patología , Displasia del Cuello del Útero/inmunología , Displasia del Cuello del Útero/patología
13.
J Neurosci Res ; 79(5): 628-37, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15668909

RESUMEN

Spinal cord injury (SCI) induces neuronal death, including apoptosis, which is completed within 24 hr at and around the impact site. We identified early proapoptotic transcriptional changes, including upregulation of proapoptotic Bax and downregulation of antiapoptotic Bcl-xL, Bcl-2, and Bcl-w, using Affymetrix DNA microarrays. Because Bcl-xL is the most robustly expressed antiapoptotic Bcl-2 molecule in adult central nervous system, we decided to characterize better the effect of SCI on Bcl-xL expression. We found Bcl-xL expressed robustly throughout uninjured spinal cord in both neurons and glia cells. We also found Bcl-xL localized in different cellular compartments: cytoplasmic, mitochondrial, and nuclear. Bcl-xL protein levels decreased in the cytoplasm and mitochondria 2 hr after SCI and persisted for 24 hr. To test the contribution of proapoptotic decreases in Bcl-xL to neuronal death, we augmented endogenous Bcl-xL levels by administering Bcl-xL fusion protein (Bcl-xL FP) into injured spinal cords. Bcl-xL FP significantly increased neuronal survival, suggesting that SCI-induced changes in Bcl-xL contribute considerably to neuronal death. Because Bcl-xL FP increases survival of dorsal horn neurons and ventral horn motoneurons, it could become clinically relevant in preserving sensory and motor functions after SCI.


Asunto(s)
Neuronas/efectos de los fármacos , Proteínas de Fusión Oncogénica/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/uso terapéutico , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Western Blotting/métodos , Recuento de Células/métodos , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/fisiología , Inmunohistoquímica/métodos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neuronas/clasificación , Neuronas/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Proteínas de Fusión Oncogénica/administración & dosificación , Fosfopiruvato Hidratasa/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Tubulina (Proteína)/metabolismo , Proteína bcl-X
15.
Am J Pathol ; 163(3): 1091-100, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12937150

RESUMEN

Nuclear envelope (NE) irregularity is an important diagnostic feature of cancer, and its molecular basis is not understood. One possible cause is abnormal postmitotic NE re-assembly, such that a rounded contour is never achieved before the next mitosis. Alternatively, dynamic forces could deform the NE during interphase following an otherwise normal postmitotic NE re-assembly. To distinguish these possibilities, normal human thyroid epithelial cells were microinjected with the papillary thyroid carcinoma oncogene (RET/PTC1 short isoform, known to induce NE irregularity), an attenuated version of RET/PTC1 lacking the leucine zipper dimerization domain (RET/PTC1 Deltazip), H (V-12) RAS, and labeled dextran. Cells were fixed at 6 or 18 to 24 hours, stained for lamins and the products of microinjected plasmids, and scored blindly using previously defined criteria for NE irregularity. 6.5% of non-injected thyrocytes showed NE irregularity. Neither dextran nor RAS microinjections increased NE irregularity. In contrast, RET/PTC1 microinjection induced NE irregularity in 27% of cells at 6 hours and 37% of cells at 18 to 24 hours. RET/PTC1 Deltazip induced significantly less irregularity. Since irregularity develops quickly, and since no mitoses and only rare possible postmitotic cells were scored, postmitotic NE re-assembly does not appear necessary for RET/PTC signaling to induce an irregular NE contour.


Asunto(s)
Interfase/fisiología , Membrana Nuclear/efectos de los fármacos , Membrana Nuclear/ultraestructura , Proteínas de Fusión Oncogénica/farmacología , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Humanos , Microinyecciones , Mitosis/fisiología , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas Tirosina Quinasas , Glándula Tiroides/efectos de los fármacos , Factores de Tiempo
16.
J Immunol ; 170(2): 861-9, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12517951

RESUMEN

Thyroid epithelial cells frequently express one or more members of the rearranged during transfection/papillary thyroid carcinoma (RET/PTC) fusion oncogene family during early stages of cancer, and fusion gene transcripts have been found in inflammatory conditions of the thyroid such as the autoimmune disease, Hashimoto's thyroiditis. Because these oncogenes encode chimeric proteins, novel RET/PTC epitopes may be targets of antitumor immune responses. We have been interested in the RET/PTC3 (RP3) fusion protein because this family member is more frequently expressed in radiation-induced and childhood papillary carcinomas than other members of the fusion oncogene family. We hypothesized that the activated kinase of c-RET, in the form of RP3, when expressed in patients with thyroid disease, presents an unusual altered self target for T cell recognition. Interestingly, we find that immunization with mouse RP3 protein can induce a strongly immunogenic response to RP3, although this response is not directed against the peptide comprising the unique fusion region. Rather, the responses are specific for the carboxyl-terminal portion of RP3 that is derived from the self protein c-RET. Furthermore, transplantation of RP3-expressing thyroid tumors into naive mice resulted in leukocytic infiltration, tumor rejection, and induction of RP3-specific T cells. Thus, the somatic fusion of two unrelated self proteins results in the development of a uniquely immunogenic response directed against self epitopes within RP3. These studies may better define the mechanisms controlling the initiation of thyroid-specific immune responses and provide insight into the design of novel molecules for invoking tumor-specific immunity.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Autoantígenos/metabolismo , Proteínas de Drosophila , Proteínas del Ojo , Proteínas de Fusión Oncogénica/biosíntesis , Neoplasias de la Tiroides/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Autoantígenos/administración & dosificación , Autoantígenos/genética , Subgrupos de Linfocitos B/inmunología , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Femenino , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Rechazo de Injerto/metabolismo , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Tolerancia Inmunológica/genética , Inmunización , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones SCID , Ratones Transgénicos , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica/administración & dosificación , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/inmunología , Biosíntesis de Proteínas , Estructura Terciaria de Proteína/genética , Proteínas/administración & dosificación , Proteínas/genética , Proteínas/fisiología , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-ret , Proteínas Tirosina Quinasas Receptoras/administración & dosificación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Subgrupos de Linfocitos T/inmunología , Glándula Tiroides/metabolismo , Glándula Tiroides/trasplante , Neoplasias de la Tiroides/genética , Tiroiditis Autoinmune/genética , Tiroiditis Autoinmune/inmunología , Vaccinia/genética
17.
Int J Hematol ; 70(4): 248-60, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10643151

RESUMEN

OBJECTIVE: All-trans-retinoic acid (ATRA), chemotherapy, and arsenic trioxide (As2O3) have been found to be effective in the treatment of acute promyelocytic leukemia (APL). Here we present a single institutional retrospective study with long-term follow-up to better define the prognostic factors and a rationale for the use of ATRA, chemotherapy, and As2O3 in the treatment of newly diagnosed and relapsed APL patients. PATIENTS AND METHODS: Newly diagnosed patients with APL entering complete remission were followed up for 3 to 95 months (n = 120). Univariate and multivariate analyses were performed to identify potential prognostic factors, including age and sex; initial white blood cell (WBC) count and peak WBC level of hyperleukocytosis during induction therapy; dose of ATRA in induction; days from induction therapy to remission; postremission therapy; type of PML-RAR alpha isoform; and follow-up of reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: The median relapse-free survival (RFS) was 26 months, and median overall survival (OS) was still not reached. The estimated 5-year RFS and OS were 34.0% +/- 6.0% and 52.5% +/- 7.9%, respectively. Initial WBC count (> or = 20 x 10(9)/l), peak level of WBC during induction, and type of postremission therapy were significantly related to survival. Our multivariate study showed that only peak level of WBC count during induction therapy and type of postremission therapy were associated with RFS and that initial WBC count was associated with OS. In relapsed patients, As2O3 was very effective and remained as the most important factor for their entering remission and survival after relapse. CONCLUSION: Through this retrospective study with long-term follow-up, some conclusions can be drawn: 1) Low-dose ATRA is as effective as the standard dose in terms of survival; 2) Initial and peak levels of WBC count during induction therapy are associated with survival; 3) A combination of chemotherapy and ATRA is better than chemotherapy or ATRA alone as postremission therapy; 4) Patients with the long form of PML-RAR alpha tend to have a more favorable OS but not RFS when compared with patients with the short form; 5) Persistent negative RT-PCR in remission is associated with favorable RFS and OS; 6) As2O3 is an effective agent for relapsed patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Promielocítica Aguda/tratamiento farmacológico , Adolescente , Adulto , Anciano , Trióxido de Arsénico , Arsenicales/administración & dosificación , Niño , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/administración & dosificación , Proteínas de Fusión Oncogénica/administración & dosificación , Óxidos/administración & dosificación , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Tretinoina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...