Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Pers Med ; 12(11)2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36422104

RESUMEN

(1) Background: The outbreak of the COVID-19 pandemic represented a real challenge for all of humanity. Characterized by a complex spectrum of signs and symptoms, by various severity degrees, the disease spread rapidly around the world. After more than two and half years since the beginning of COVID-19 pandemic, in the context of a paradoxical, enigmatic, and relentless COVID-19, the objective of the current study was to evaluate the characteristics and evolution of patients with SARS-CoV-2 infection, hospitalized in "St. Parascheva" Clinical Hospital of Infectious Diseases (Iasi, Romania). (2) Methods: This is a retrospective study that used the medical database recorded between July and November 2021 in order to highlight the characteristics of SARS-CoV-2 infection in patients from the northeastern region of Romania. (3) Results: We enrolled in the study a total of 1732 SARS-CoV-2 infected patients, mean age 67 ± 3.4 years, the female gender predominating (987 cases; 56.98%) as well as patients from the urban environment (982 patients; 56.69%). Moderate form of the disease predominated (814 cases; 47%), pulmonary imaging changes were found in 1042 (60.16%) cases, and 1242 (71.71%) patients had at least one underlying disease. After a median length of hospitalization of 9.5 days, 1359 (78.46%) patients were discharged cured, 48 (2.77%) were transferred to other services by decompensating the associated pathologies, 302 (17.43%) patients needed extensive support in the intensive care unit and there were 325 (18.76%) deaths. (4) Conclusions: The epidemiological characteristics of SARS-CoV-2 infection recorded in our study were mostly the same as characteristics of COVID-19 from all over the world.

2.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33771892

RESUMEN

BACKGROUND: The cancer-testis antigen MAGE-A4 is an attractive target for T-cell-based immunotherapy, especially for indications with unmet clinical need like non-small cell lung or triple-negative breast cancer. METHODS: An unbiased CD137-based sorting approach was first used to identify an immunogenic MAGE-A4-derived epitope (GVYDGREHTV) that was properly processed and presented on human leukocyte antigen (HLA)-A2 molecules encoded by the HLA-A*02:01 allele. To isolate high-avidity T cells via subsequent multimer sorting, an in vitro priming approach using HLA-A2-negative donors was conducted to bypass central tolerance to this self-antigen. Pre-clinical parameters of safety and activity were assessed in a comprehensive set of in vitro and in vivo studies. RESULTS: A MAGE-A4-reactive, HLA-A2-restricted T-cell receptor (TCR) was isolated from primed T cells of an HLA-A2-negative donor. The respective TCR-T-cell (TCR-T) product bbT485 was demonstrated pre-clinically to have a favorable safety profile and superior in vivo potency compared with TCR-Ts expressing a TCR derived from a tolerized T-cell repertoire to self-antigens. This natural high-avidity TCR was found to be CD8 co-receptor independent, allowing effector functions to be elicited in transgenic CD4+ T helper cells. These CD4+ TCR-Ts supported an anti-tumor response by direct killing of MAGE-A4-positive tumor cells and upregulated hallmarks associated with helper function, such as CD154 expression and release of key cytokines on tumor-specific stimulation. CONCLUSION: The extensive pre-clinical assessment of safety and in vivo potency of bbT485 provide the basis for its use in TCR-T immunotherapy studies. The ability of this non-mutated high-avidity, co-receptor-independent TCR to activate CD8+ and CD4+ T cells could potentially provide enhanced cellular responses in the clinical setting through the induction of functionally diverse T-cell subsets that goes beyond what is currently tested in the clinic.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos CD8/inmunología , Linfocitos T CD8-positivos/trasplante , Inmunoterapia Adoptiva , Proteínas de Neoplasias/inmunología , Neoplasias/terapia , Receptores Quiméricos de Antígenos/inmunología , Células A549 , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Células HEK293 , Antígeno HLA-A2/inmunología , Antígeno HLA-A2/metabolismo , Humanos , Epítopos Inmunodominantes , Células K562 , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Carga Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Clin Cancer Res ; 25(24): 7340-7350, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31558475

RESUMEN

PURPOSE: Current protocols for CD19 chimeric antigen receptor-expressing T cells (CD19.CAR-T cells) require recipients to tolerate preinfusion cytoreductive chemotherapy, and the presence of sufficient target antigen on normal or malignant B cells. PATIENTS AND METHODS: We investigated whether additional stimulation of CD19.CAR-T cells through their native receptors can substitute for cytoreductive chemotherapy, inducing expansion and functional persistence of CD19.CAR-T even in patients in remission of B-cell acute lymphocytic leukemia. We infused a low dose of CD19.CAR-modified virus-specific T cells (CD19.CAR-VST) without prior cytoreductive chemotherapy into 8 patients after allogeneic stem cell transplant. RESULTS: Absent virus reactivation, we saw no CD19.CAR-VST expansion. In contrast, in patients with viral reactivation, up to 30,000-fold expansion of CD19.CAR-VSTs was observed, with depletion of CD19+ B cells. Five patients remain in remission at 42-60+ months. CONCLUSIONS: Dual T-cell receptor and CAR stimulation can thus potentiate effector cell expansion and CAR-target cell killing, even when infusing low numbers of effector cells without cytoreduction.


Asunto(s)
Antígenos CD19/inmunología , Inmunoterapia Adoptiva/métodos , Linfoma de Células B/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T/trasplante , Adenoviridae/fisiología , Adolescente , Antígenos CD19/metabolismo , Niño , Preescolar , Vectores Genéticos , Herpesvirus Humano 4/fisiología , Humanos , Linfoma de Células B/inmunología , Linfoma de Células B/virología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/virología , Retroviridae/fisiología , Linfocitos T/inmunología , Linfocitos T/virología , Adulto Joven
4.
Mol Ther ; 25(3): 580-592, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28187946

RESUMEN

Immunotherapy with T cells expressing the chimeric antigen receptor (CAR) specific for the CD19 antigen (CD19.CAR-Ts) is a very effective treatment in B cell lymphoid malignancies. However, B cell aplasia and cytokine release syndrome (CRS) secondary to the infusion of CD19.CAR-Ts remain significant drawbacks. The inclusion of safety switches into the vector encoding the CAR is seen as the safest method to terminate the effects of CD19.CAR-Ts in case of severe toxicities or after achieving long-term sustained remissions. By contrast, the complete elimination of CD19.CAR-Ts when CRS occurs may jeopardize clinical responses as CRS and antitumor activity seem to concur. We have demonstrated, in a humanized mouse model, that the inducible caspase-9 (iC9) safety switch can eliminate CD19.CAR-Ts in a dose-dependent manner, allowing either a selective containment of CD19.CAR-T expansion in case of CRS or complete deletion on demand granting normal B cell reconstitution.


Asunto(s)
Antígenos CD19/inmunología , Caspasa 9/metabolismo , Citotoxicidad Inmunológica , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Apoptosis/genética , Apoptosis/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proliferación Celular , Supervivencia Celular , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Humanos , Inmunoterapia Adoptiva , Activación de Linfocitos/inmunología , Ratones , Imagen Molecular , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/terapia , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos T/genética
5.
Cancer Immunol Immunother ; 65(6): 741-51, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27129972

RESUMEN

Neutrophil elastase (NE) is an innate immune cell-derived inflammatory mediator that we have shown increases the presentation of tumor-associated peptide antigens in breast cancer. In this study, we extend these observations to show that NE uptake has a broad effect on enhancing antigen presentation by breast cancer cells. We show that NE increases human leukocyte antigen (HLA) class I expression on the surface of breast cancer cells in a concentration and time-dependent manner. HLA class I upregulation requires internalization of enzymatically active NE. Western blots of NE-treated breast cancer cells confirm that the expression of total HLA class I as well as the antigen-processing machinery proteins TAP1, LMP2, and calnexin does not change following NE treatment. This suggests that NE does not increase the efficiency of antigen processing; rather, it mediates the upregulation of HLA class I by stabilizing and reducing membrane recycling of HLA class I molecules. Furthermore, the effects of NE extend beyond breast cancer since the uptake of NE by EBV-LCL increases the presentation of HLA class I-restricted viral peptides, as shown by their increased sensitivity to lysis by EBV-specific CD8+ T cells. Together, our results show that NE uptake increases the responsiveness of breast cancer cells to adaptive immunity by broad upregulation of membrane HLA class I and support the conclusion that the innate inflammatory mediator NE enhances tumor cell recognition and increases tumor sensitivity to the host adaptive immune response.


Asunto(s)
Presentación de Antígeno/inmunología , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase I/genética , Elastasa de Leucocito/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Péptidos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
6.
Mol Ther ; 24(1): 175-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26310629

RESUMEN

Despite many clinical trials conducted with oncolytic viruses, the exact tumor-level mechanisms affecting therapeutic efficacy have not been established. Currently there are no biomarkers available that would predict the clinical outcome to any oncolytic virus. To assess the baseline immunological phenotype and find potential prognostic biomarkers, we monitored mRNA expression levels in 31 tumor biopsy or fluid samples from 27 patients treated with oncolytic adenovirus. Additionally, protein expression was studied from 19 biopsies using immunohistochemical staining. We found highly significant changes in several signaling pathways and genes associated with immune responses, such as B-cell receptor signaling (P < 0.001), granulocyte macrophage colony-stimulating factor (GM-CSF) signaling (P < 0.001), and leukocyte extravasation signaling (P < 0.001), in patients surviving a shorter time than their controls. In immunohistochemical analysis, markers CD4 and CD163 were significantly elevated (P = 0.020 and P = 0.016 respectively), in patients with shorter than expected survival. Interestingly, T-cell exhaustion marker TIM-3 was also found to be significantly upregulated (P = 0.006) in patients with poor prognosis. Collectively, these data suggest that activation of several functions of the innate immunity before treatment is associated with inferior survival in patients treated with oncolytic adenovirus. Conversely, lack of chronic innate inflammation at baseline may predict improved treatment outcome, as suggested by good overall prognosis.


Asunto(s)
Adenoviridae/fisiología , Perfilación de la Expresión Génica/métodos , Inmunidad Innata , Neoplasias/genética , Neoplasias/terapia , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Antígenos CD4/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias/inmunología , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Pronóstico , Receptores de Superficie Celular/metabolismo , Resultado del Tratamiento
7.
Oncotarget ; 6(31): 30675-703, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26362400

RESUMEN

The emergence of genetic engineering at the beginning of the 1970's opened the era of biomedical technologies, which aims to improve human health using genetic manipulation techniques in a clinical context. Gene therapy represents an innovating and appealing strategy for treatment of human diseases, which utilizes vehicles or vectors for delivering therapeutic genes into the patients' body. However, a few past unsuccessful events that negatively marked the beginning of gene therapy resulted in the need for further studies regarding the design and biology of gene therapy vectors, so that this innovating treatment approach can successfully move from bench to bedside. In this paper, we review the major gene delivery vectors and recent improvements made in their design meant to overcome the issues that commonly arise with the use of gene therapy vectors. At the end of the manuscript, we summarized the main advantages and disadvantages of common gene therapy vectors and we discuss possible future directions for potential therapeutic vectors.


Asunto(s)
Técnicas de Transferencia de Gen , Ingeniería Genética/métodos , Terapia Genética/métodos , Vectores Genéticos/genética , Adenoviridae/genética , Dependovirus/genética , Humanos , Lentivirus/genética , Plásmidos/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , Retroviridae/genética , Replicación Viral/genética
8.
Hum Gene Ther ; 26(3): 134-44, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25557131

RESUMEN

For long it has been recognized that tumor necrosis factor alpha (TNFa) has anticancer characteristics, and its use as a cancer therapeutic was proposed already in the 1980s. However, its systemic toxicity has limited its usability. Oncolytic viruses, selectively cancer-killing viruses, have shown great potency, and one of their most useful aspects is their ability to produce high amounts of transgene products locally, resulting in high local versus systemic concentrations. Therefore, the overall magnitude of tumor cell killing results from the combination of oncolysis, transgene-mediated direct effect such as TNFa-mediated apoptosis, and, perhaps most significantly, from activation of the host immune system against the tumor. We generated a novel chimeric oncolytic adenovirus expressing human TNFa, Ad5/3-D24-hTNFa, whose efficacy and immunogenicity were tested in vitro and in vivo. The hTNFa-expressing adenovirus showed increased cancer-eradicating potency, which was shown to be because of elevated apoptosis and necrosis rates and induction of various immune responses. Interestingly, we saw increase in immunogenic cell death markers in Ad5/3-d24-hTNFa-treated cells. Moreover, tumors treated with Ad5/3-D24-hTNFa displayed enhanced presence of OVA-specific cytotoxic T cells. We thus can conclude that tumor eradication and antitumor immune responses mediated by Ad5/3-d24-hTNFa offer a new potential drug candidate for cancer therapy.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Neoplasias/genética , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Factor de Necrosis Tumoral alfa/metabolismo , Apoptosis/inmunología , Línea Celular Tumoral , Vectores Genéticos/inmunología , Células HEK293 , Humanos , Neoplasias/inmunología , Estadísticas no Paramétricas , Linfocitos T Citotóxicos/inmunología , Factor de Necrosis Tumoral alfa/genética
9.
Int J Cancer ; 136(5): 1065-72, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25042001

RESUMEN

Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Viroterapia Oncolítica , Neoplasias Pancreáticas/inmunología , Virus Vaccinia/genética , Replicación Viral/inmunología , Animales , Movimiento Celular , Proliferación Celular , Células Cultivadas , Chlorocebus aethiops , Técnicas de Cocultivo , Cricetinae , ADN Viral/genética , Humanos , Técnicas para Inmunoenzimas , Macrófagos , Mesocricetus , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T , Virus Vaccinia/inmunología , Células Vero , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Semin Oncol ; 41(5): 661-6, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25440610

RESUMEN

Monoclonal antibodies (mAbs) and their directly derived cell-based application known as chimeric antigen receptors (CARs) ensue from the need to develop novel therapeutic strategies that retain high anti-tumor activity, but carry reduced toxicity compared to conventional chemo- and radiotherapies. In this concise review article, we will summarize the application of antibodies designed to target antigens expressed by tumor cells, and the transition from these antibodies to the generation of CARs.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Genes Codificadores de los Receptores de Linfocitos T/inmunología , Neoplasias/terapia , Receptores de Antígenos/inmunología , Receptores de Antígenos/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Linfocitos T/inmunología
11.
Cancer Res ; 74(18): 5195-205, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25060519

RESUMEN

The clinical efficacy of chimeric antigen receptor (CAR)-redirected T cells remains marginal in solid tumors compared with leukemias. Failures have been attributed to insufficient T-cell migration and to the highly immunosuppressive milieu of solid tumors. To overcome these obstacles, we have combined CAR-T cells with an oncolytic virus armed with the chemokine RANTES and the cytokine IL15, reasoning that the modified oncolytic virus will both have a direct lytic effect on infected malignant cells and facilitate migration and survival of CAR-T cells. Using neuroblastoma as a tumor model, we found that the adenovirus Ad5Δ24 exerted a potent, dose-dependent, cytotoxic effect on tumor cells, whereas CAR-T cells specific for the tumor antigen GD2 (GD2.CAR-T cells) were not damaged. When used in combination, Ad5Δ24 directly accelerated the caspase pathways in tumor cells exposed to CAR-T cells, whereas the intratumoral release of both RANTES and IL15 attracted CAR-T cells and promoted their local survival, respectively, increasing the overall survival of tumor-bearing mice. These preclinical data support the use of this innovative biologic platform of immunotherapy for solid tumors. Cancer Res; 74(18); 5195-205. ©2014 AACR.


Asunto(s)
Inmunoterapia/métodos , Neuroblastoma/inmunología , Neuroblastoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Linfocitos T/inmunología , Animales , Línea Celular Tumoral , Citocinas/inmunología , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos NOD , Microscopía Confocal , Neuroblastoma/virología , Receptores de Antígenos de Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Cancer ; 135(3): 720-30, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24374597

RESUMEN

Sarcomas are a relatively rare cancer, but often incurable at the late metastatic stage. Oncolytic immunotherapy has gained attention over the past years, and a wide range of oncolytic viruses have been delivered via intratumoral injection with positive safety and promising efficacy data. Here, we report preclinical and clinical results from treatment of sarcoma with oncolytic adenovirus Ad5/3-D24-GMCSF (CGTG-102). Ad5/3-D24-GMCSF is a serotype chimeric oncolytic adenovirus coding for human granulocyte-macrophage colony-stimulating factor (GM-CSF). The efficacy of Ad5/3-D24-GMCSF was evaluated on a panel of soft-tissue sarcoma (STS) cell lines and in two animal models. Sarcoma specific human data were also collected from the Advanced Therapy Access Program (ATAP), in preparation for further clinical development. Efficacy was seen in both in vitro and in vivo STS models. Fifteen patients with treatment-refractory STS (13/15) or primary bone sarcoma (2/15) were treated in ATAP, and treatments appeared safe and well-tolerated. A total of 12 radiological RECIST response evaluations were performed, and two cases of minor response, six cases of stable disease and four cases of progressive disease were detected in patients progressing prior to virus treatment. Overall, the median survival time post treatment was 170 days. One patient is still alive at 1,459 days post virus treatment. In summary, Ad5/3-D24-GMCSF appears promising for the treatment of advanced STS; a clinical trial for treatment of refractory injectable solid tumors including STS is ongoing.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Viroterapia Oncolítica , Sarcoma/terapia , Animales , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Inyecciones Intralesiones , Mesocricetus , Ratones , Ratones Desnudos , Pronóstico , Sarcoma/sangre , Sarcoma/mortalidad , Tasa de Supervivencia , Células Tumorales Cultivadas , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Clin Chim Acta ; 425: 128-38, 2013 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-23933123

RESUMEN

During the last decades the incidence of cancer increased dramatically especially in developed countries. In spite of the fact that the immunochemical methods allowed the diagnosis in early stages, the biopsies are generally invasive methods that create discomfort to patients. The need for fast, sensitive, easy to use and noninvasive diagnosis tools is actually of great interest for many research groups all over the world. Immunosensors (ISs) are miniaturized measuring devices, which selectively detect their targets by means of antibodies (Abs) and provide concentration-dependent signals. Ab binding leads to a variation in electric charge, mass, heat or optical properties, which can be detected directly or indirectly by a variety of transducers. A great number of proteins could be considered as recognition element. In this review the attention was focused on main cancer biomarkers, currently used by immunological methods (immunohistochemistry, ELISA, flow cytometry, Western blot, immunofluorescence etc) and in the development of electrochemical immunoassays that could be used in cancer diagnosis, prognosis and therapy monitoring.


Asunto(s)
Biomarcadores de Tumor/sangre , Técnicas Biosensibles/instrumentación , Neoplasias de la Mama/sangre , Técnicas Electroquímicas , Inmunoensayo/instrumentación , Neoplasias Ováricas/sangre , Anticuerpos/sangre , Proteína BRCA1/sangre , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/inmunología , Antígeno Carcinoembrionario/sangre , Femenino , Humanos , Mucina-1/sangre , Mucinas/sangre , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/inmunología , Proteínas/metabolismo , Receptor ErbB-2/sangre , Proteína 2 de Dominio del Núcleo de Cuatro Disulfuros WAP
14.
Mol Ther ; 21(6): 1212-23, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23546299

RESUMEN

Oncolytic adenoviruses and certain chemotherapeutics can induce autophagy and immunogenic cancer cell death. We hypothesized that the combination of oncolytic adenovirus with low-dose temozolomide (TMZ) is safe, effective, and capable of inducing antitumor immune responses. Metronomic low-dose cyclophosphamide (CP) was added to selectively reduce regulatory T-cells. Preclinically, combination therapy inhibited tumor growth, increased autophagy, and triggered immunogenic cell death as indicated by elevated calreticulin, adenosine triphosphate (ATP) release, and nuclear protein high-mobility group box-1 (HMGB1) secretion. A total of 41 combination treatments given to 17 chemotherapy-refractory cancer patients were well tolerated. We observed anti- and proinflammatory cytokine release, evidence of virus replication, and induction of neutralizing antibodies. Tumor cells showed increased autophagy post-treatment. Release of HMGB1 into serum--a possible indicator of immune response--increased in 60% of treatments, and seemed to correlate with tumor-specific T-cell responses, observed in 10/15 cases overall (P = 0.0833). Evidence of antitumor efficacy was seen in 67% of evaluable treatments with a trend for increased survival over matched controls treated with virus only. In summary, the combination of oncolytic adenovirus with low-dose TMZ and metronomic CP increased tumor cell autophagy, elicited antitumor immune responses, and showed promising safety and efficacy.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Dacarbazina/análogos & derivados , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Adenosina Trifosfato/metabolismo , Adenoviridae/fisiología , Adolescente , Adulto , Anciano , Animales , Anticuerpos Neutralizantes/sangre , Calreticulina/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Niño , Terapia Combinada/métodos , Ciclofosfamida/farmacología , Citocinas/sangre , ADN Viral/sangre , Dacarbazina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Proteína HMGB1/sangre , Proteína HMGB1/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Microscopía Electrónica , Persona de Mediana Edad , Virus Oncolíticos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Temozolomida , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
15.
Clin Cancer Res ; 19(10): 2734-44, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23493351

RESUMEN

PURPOSE: Multiple injections of oncolytic adenovirus could enhance immunologic response. In the first part of this article, the focus was on immunologic aspects. Sixty patients previously naïve to oncolytic virus and who had white blood cells available were treated. Thirty-nine of 60 were assessed after a single virus administration, whereas 21 of 60 received a "serial treatment" consisting of three injections within 10 weeks. In the second part, we focused on 115 patients treated with a granulocyte macrophage colony-stimulating factor (GM-CSF)-coding capsid chimeric adenovirus, CGTG-102. RESULTS: Following serial treatment, both increase and decrease in antitumor T cells in blood were seen more frequently, findings which are compatible with induction of T-cell immunity and trafficking of T cells to tumors, respectively. Safety was good in both groups. In 115 patients treated with CGTG-102 (Ad5/3-D24-GMCSF), median overall survival was 111 days following single and 277 days after serial treatment in nonrandomized comparison. Switching the virus capsid for avoiding neutralizing antibodies in a serial treatment featuring three different viruses did not impact safety or efficacy. A correlation between antiviral and antitumor T cells was seen (P = 0.001), suggesting that viral oncolysis can result in epitope spreading and breaking of tumor-associated immunologic tolerance. Alternatively, some patients may be more susceptible to induction of T-cell immunity and/or trafficking. CONCLUSIONS: These results provide the first human data linking antiviral immunity with antitumor immunity, implying that oncolytic viruses could have an important role in cancer immunotherapy.


Asunto(s)
Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Linfocitos T/inmunología , Adenoviridae/genética , Adenoviridae/inmunología , Adolescente , Adulto , Anciano , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Niño , Anergia Clonal/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-10/inmunología , Interleucina-10/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/clasificación , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Análisis de Supervivencia , Linfocitos T/metabolismo , Factores de Tiempo , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Adulto Joven
16.
Mol Ther ; 20(9): 1821-30, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22871667

RESUMEN

Twenty-five patients with chemotherapy refractory cancer were treated with a fully serotype 3-based oncolytic adenovirus Ad3-hTERT-E1A. In mice, Ad3 induced higher amounts of cytokines but less liver damage than Ad5 or Ad5/3. In humans, the only grade 3 adverse reactions were self-limiting cytopenias and generally the safety profile resembled Ad5-based oncolytic viruses. Patients that had been previously treated with Ad5 viruses presented longer lasting lymphocytopenia but no median increase in Ad3-specific T-cells in blood, suggesting immunological activity against antigens other than Ad3 hexon. Frequent alterations in antitumor T-cells in blood were seen regardless of previous virus exposure. Neutralizing antibodies against Ad3 increased in all patients, whereas Ad5 neutralizing antibodies remained stable. Treatment with Ad3-hTERT-E1A resulted in re-emergence of Ad5 viruses from previous treatments into blood and vice versa. Signs of possible efficacy were seen in 11/15 (73%) patients evaluable for tumor markers, four of which were treated only intravenously. Particularly promising results were seen in breast cancer patients and especially those receiving concomitant trastuzumab. Taken together, Ad3-hTERT-E1A seems safe for further clinical testing or development of armed versions. It offers an immunologically attractive alternative, with possible pharmacodynamic differences and a different receptor compared to Ad5.


Asunto(s)
Adenoviridae/inmunología , Terapia Genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/inmunología , Adenoviridae/genética , Adulto , Anciano , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/genética , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/terapia , Resistencia a Antineoplásicos , Femenino , Genes Virales , Vectores Genéticos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/mortalidad , Virus Oncolíticos/genética , Tasa de Supervivencia , Linfocitos T/inmunología , Trastuzumab , Replicación Viral
17.
Mol Ther ; 20(11): 2076-86, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22828500

RESUMEN

Oncolytic viruses represent a multifaceted tool for cancer treatment. In addition to specific killing of cancer cells (oncolysis), these agents also provide danger signals prompting the immune system to stimulate an antitumor immune response. To increase adenovirus adjuvancy, we engineered the genome of Ad5D24 by inserting 18 immunostimulatory islands (Ad5D24-CpG). The toxicity and immunogenicity profile of Ad5D24-CpG showed that the safety of the maternal virus was retained. The efficacy of the CpG-enriched virus was assessed in a xenograft model of lung cancer where a significant increase in antitumor effect was seen in comparison with controls. When the experiment was repeated in animal depleted of natural killer (NK) cells, Ad5D24-CpG lost its advantage. The same was seen when Toll-like receptor (TLR)9 was blocked systemically. In a syngeneic model of melanoma (B16-OVA), we observed a significant increase of OVA-specific T cells and a decrease of activation of myeloid-derived suppressor cells in Ad5D24-CpG-treated mice. In conclusion, we have generated the first genetically modified oncolytic adenovirus backbone able to enhance TLR9-stimulation for increased antitumor activity.


Asunto(s)
Adenoviridae/genética , Neoplasias Pulmonares/terapia , Melanoma/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Receptor Toll-Like 9/agonistas , Adenoviridae/inmunología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular , Terapia Combinada , Islas de CpG/inmunología , Células HEK293 , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Melanoma/inmunología , Melanoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células Mieloides , FN-kappa B/metabolismo , Oligodesoxirribonucleótidos/farmacología , Oligodesoxirribonucleótidos/uso terapéutico , Virus Oncolíticos/inmunología , Receptor Toll-Like 9/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 72(9): 2327-38, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22396493

RESUMEN

Oncolytic adenovirus is an attractive platform for immunotherapy because virus replication is highly immunogenic and not subject to tolerance. Although oncolysis releases tumor epitopes and provides costimulatory danger signals, arming the virus with immunostimulatory molecules can further improve efficacy. CD40 ligand (CD40L, CD154) induces apoptosis of tumor cells and triggers several immune mechanisms, including a T-helper type 1 (T(H)1) response, which leads to activation of cytotoxic T cells and reduction of immunosuppression. In this study, we constructed a novel oncolytic adenovirus, Ad5/3-hTERT-E1A-hCD40L, which features a chimeric Ad5/3 capsid for enhanced tumor transduction, a human telomerase reverse transcriptase (hTERT) promoter for tumor selectivity, and human CD40L for increased efficacy. Ad5/3-hTERT-E1A-hCD40L significantly inhibited tumor growth in vivo via oncolytic and apoptotic effects, and (Ad5/3-hTERT-E1A-hCD40L)-mediated oncolysis resulted in enhanced calreticulin exposure and HMGB1 and ATP release, which were suggestive of immunogenicity. In two syngeneic mouse models, murine CD40L induced recruitment and activation of antigen-presenting cells, leading to increased interleukin-12 production in splenocytes. This effect was associated with induction of the T(H)1 cytokines IFN-γ, RANTES, and TNF-α. Tumors treated with Ad5/3-CMV-mCD40L also displayed an enhanced presence of macrophages and cytotoxic CD8(+) T cells but not B cells. Together, our findings show that adenoviruses coding for CD40L mediate multiple antitumor effects including oncolysis, apoptosis, induction of T-cell responses, and upregulation of T(H)1 cytokines.


Asunto(s)
Ligando de CD40/genética , Ligando de CD40/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Animales , Apoptosis/inmunología , Línea Celular Tumoral , Citocinas/inmunología , Humanos , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias/genética , Neoplasias/virología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/terapia , Neoplasias de la Vejiga Urinaria/virología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 7(3): e32871, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22412937

RESUMEN

Oncolytic adenoviruses can be engineered for better tumor selectivity, gene delivery and be armed for imaging and concentrating radionuclides into tumors for synergistic oncolysis. We constructed Ad5/3-hTERT-hNIS where replication is controlled by hTERT-promoter. Ad5/3-hTERT-hNIS expresses hNIS for imaging of transgene expression and for treatment of infected tumors by radioiodine. Ad5/3-hTERT-hNIS efficiently killed prostate cancer cells and induced iodine uptake in vitro and in vivo after intratumoral virus administration. Survival of mice treated with intravenous Ad5/3-hTERT-hNIS significantly prolonged survival over mock or radioiodine only but the combination of virus with radioiodine was not more effective than virus alone. Temporal and spatial changes in hNIS-expression during therapy were detected with SPECT, demonstrating feasibility of evaluation of the combination therapy with hNIS-expressing adenoviruses and radioiodide.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/genética , Radioisótopos de Yodo/metabolismo , Imagen Multimodal , Virus Oncolíticos/genética , Tomografía de Emisión de Positrones , Simportadores/genética , Tomografía Computarizada por Rayos X , Animales , Línea Celular , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Inyecciones Intravenosas , Radioisótopos de Yodo/uso terapéutico , Masculino , Ratones , Ratones Desnudos , Imagen Molecular , Viroterapia Oncolítica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/terapia , Simportadores/metabolismo , Resultado del Tratamiento
20.
Cancer Res ; 72(7): 1621-31, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22323527

RESUMEN

The immunosuppressive environment of advanced tumors is a primary obstacle to the efficacy of immunostimulatory and vaccine approaches. Here, we report an approach to arm an oncolytic virus with CD40 ligand (CD40L) to stimulate beneficial immunologic responses in patients. A double-targeted chimeric adenovirus controlled by the hTERT promoter and expressing CD40L (CGTG-401) was constructed and nine patients with progressing advanced solid tumors refractory to standard therapies were treated intratumorally. No serious adverse events resulting in patient hospitalization occurred. Moderate or no increases in neutralizing antibodies were seen, suggesting effective Th1 immunologic effects. An assessment of the blood levels of virus indicated 17.5% of the samples (n = 40) were positive at a low level early after treatment, but not thereafter. In contrast, high levels of virus, CD40L, and RANTES were documented locally at the tumor. Peripheral blood mononuclear cells were analyzed by IFN-γ ELISPOT analysis and induction of both survivin-specific and adenovirus-specific T cells was seen. Antitumor T-cell responses were even more pronounced when assessed by intracellular cytokine staining after stimulation with tumor type-specific peptide pools. Of the evaluable patients, 83% displayed disease control at 3 months and in both cases in which treatment was continued the effect was sustained for at least 8 months. Injected and noninjected lesions responded identically. Together, these findings support further clinical evaluation of CGTG-401.


Asunto(s)
Adenoviridae/genética , Ligando de CD40/genética , Inmunoterapia , Neoplasias/terapia , Viroterapia Oncolítica , Adenoviridae/inmunología , Adulto , Anticuerpos Antivirales/análisis , Ligando de CD40/análisis , Quimiocina CCL5/análisis , Femenino , Humanos , Interferón gamma/biosíntesis , Masculino , Persona de Mediana Edad , Imagen Multimodal , Neoplasias/inmunología , Viroterapia Oncolítica/efectos adversos , Tomografía de Emisión de Positrones , Telomerasa/genética , Células TH1/inmunología , Tomografía Computarizada por Rayos X , Virión/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...