Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 14(9)2022 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-35565310

RESUMEN

Pexa-Vec is an engineered Wyeth-strain vaccinia oncolytic virus (OV), which has been tested extensively in clinical trials, demonstrating enhanced cytotoxic T cell infiltration into tumours following treatment. Favourable immune consequences to Pexa-Vec include the induction of an interferon (IFN) response, followed by inflammatory cytokine/chemokine secretion. This promotes tumour immune infiltration, innate and adaptive immune cell activation and T cell priming, culminating in targeted tumour cell killing, i.e., an immunologically 'cold' tumour microenvironment is transformed into a 'hot' tumour. However, as with all immunotherapies, not all patients respond in a uniformly favourable manner. Our study herein, shows a differential immune response by patients to intravenous Pexa-Vec therapy, whereby some patients responded to the virus in a typical and expected manner, demonstrating a significant IFN induction and subsequent peripheral immune activation. However, other patients experienced a markedly subdued immune response and appeared to exhibit an exhausted phenotype at baseline, characterised by higher baseline immune checkpoint expression and regulatory T cell (Treg) levels. This differential baseline immunological profile accurately predicted the subsequent response to Pexa-Vec and may, therefore, enable the development of predictive biomarkers for Pexa-Vec and OV therapies more widely. If confirmed in larger clinical trials, these immunological biomarkers may enable a personalised approach, whereby patients with an exhausted baseline immune profile are treated with immune checkpoint blockade, with the aim of reversing immune exhaustion, prior to or alongside OV therapy.

2.
Cancer Immunol Res ; 10(6): 745-756, 2022 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-35439304

RESUMEN

Improving the chances of curing patients with cancer who have had surgery to remove metastatic sites of disease is a priority area for cancer research. Pexa-Vec (Pexastimogene Devacirepvec; JX-594, TG6006) is a principally immunotherapeutic oncolytic virus that has reached late-phase clinical trials. We report the results of a single-center, nonrandomized biological end point study (trial registration: EudraCT number 2012-000704-15), which builds on the success of the presurgical intravenous delivery of oncolytic viruses to tumors. Nine patients with either colorectal cancer liver metastases or metastatic melanoma were treated with a single intravenous infusion of Pexa-Vec ahead of planned surgical resection of the metastases. Grade 3 and 4 Pexa-Vec-associated side effects were lymphopaenia and neutropaenia. Pexa-Vec was peripherally carried in plasma and was not associated with peripheral blood mononuclear cells. Upon surgical resection, Pexa-Vec was found in the majority of analyzed tumors. Pexa-Vec therapy associated with IFNα secretion, chemokine induction, and resulted in transient innate and long-lived adaptive anticancer immunity. In the 2 patients with significant and complete tumor necrosis, a reduction in the peripheral T-cell receptor diversity was observed at the time of surgery. These results support the development of presurgical oncolytic vaccinia virus-based therapies to stimulate anticancer immunity and increase the chances to cure patients with cancer.


Asunto(s)
Neoplasias Hepáticas , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Leucocitos Mononucleares , Neoplasias Hepáticas/terapia , Terapia Neoadyuvante , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Vaccinia/genética
3.
J Immunother Cancer ; 7(1): 164, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31262361

RESUMEN

BACKGROUND: The oncolytic virus, coxsackievirus A21 (CVA21), has shown promise as a single agent in several clinical trials and is now being tested in combination with immune checkpoint blockade. Combination therapies offer the best chance of disease control; however, the design of successful combination strategies requires a deeper understanding of the mechanisms underpinning CVA21 efficacy, in particular, the role of CVA21 anti-tumor immunity. Therefore, this study aimed to examine the ability of CVA21 to induce human anti-tumor immunity, and identify the cellular mechanism responsible. METHODS: This study utilized peripheral blood mononuclear cells from i) healthy donors, ii) Acute Myeloid Leukemia (AML) patients, and iii) patients taking part in the STORM clinical trial, who received intravenous CVA21; patients receiving intravenous CVA21 were consented separately in accordance with local institutional ethics review and approval. Collectively, these blood samples were used to characterize the development of innate and adaptive anti-tumor immune responses following CVA21 treatment. RESULTS: An Initial characterization of peripheral blood mononuclear cells, collected from cancer patients following intravenous infusion of CVA21, confirmed that CVA21 activated immune effector cells in patients. Next, using hematological disease models which were sensitive (Multiple Myeloma; MM) or resistant (AML) to CVA21-direct oncolysis, we demonstrated that CVA21 stimulated potent anti-tumor immune responses, including: 1) cytokine-mediated bystander killing; 2) enhanced natural killer cell-mediated cellular cytotoxicity; and 3) priming of tumor-specific cytotoxic T lymphocytes, with specificity towards known tumor-associated antigens. Importantly, immune-mediated killing of both MM and AML, despite AML cells being resistant to CVA21-direct oncolysis, was observed. Upon further examination of the cellular mechanisms responsible for CVA21-induced anti-tumor immunity we have identified the importance of type I IFN for NK cell activation, and demonstrated that both ICAM-1 and plasmacytoid dendritic cells were key mediators of this response. CONCLUSION: This work supports the development of CVA21 as an immunotherapeutic agent for the treatment of both AML and MM. Additionally, the data presented provides an important insight into the mechanisms of CVA21-mediated immunotherapy to aid the development of clinical biomarkers to predict response and rationalize future drug combinations.


Asunto(s)
Enterovirus , Leucemia Mieloide Aguda/terapia , Viroterapia Oncolítica , Virus Oncolíticos , Inmunidad Adaptativa , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular Tumoral , Células Dendríticas/inmunología , Femenino , Humanos , Inmunidad Innata , Molécula 1 de Adhesión Intercelular/inmunología , Leucemia Mieloide Aguda/inmunología , Masculino , Persona de Mediana Edad , Linfocitos T Citotóxicos/inmunología
4.
Mol Ther ; 27(6): 1139-1152, 2019 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-31053413

RESUMEN

A clinical oncolytic herpes simplex virus (HSV) encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), talimogene laherparepvec, causes regression of injected and non-injected melanoma lesions in patients and is now licensed for clinical use in advanced melanoma. To date, limited data are available regarding the mechanisms of human anti-tumor immune priming, an improved understanding of which could inform the development of future combination strategies with improved efficacy. This study addressed direct oncolysis and innate and adaptive human immune-mediated effects of a closely related HSV encoding GM-CSF (HSVGM-CSF) alone and in combination with histone deacetylase inhibition. We found that HSVGM-CSF supported activation of anti-melanoma immunity via monocyte-mediated type I interferon production, which activates NK cells, and viral maturation of immature dendritic cells (iDCs) into potent antigen-presenting cells for cytotoxic T lymphocyte (CTL) priming. Addition of the histone deacetylase inhibitor valproic acid (VPA) to HSVGM-CSF treatment of tumor cells increased viral replication, viral GM-CSF production, and oncolysis and augmented the development of anti-tumor immunity. Mechanistically, VPA increased expression of activating ligands for NK cell recognition and induced expression of tumor-associated antigens, supporting innate NK cell killing and CTL priming. These data support the clinical combination of talimogene laherparepvec with histone deacetylase inhibition to enhance oncolysis and anti-tumor immunity.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Melanoma/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Simplexvirus/inmunología , Neoplasias Cutáneas/terapia , Ácido Valproico/uso terapéutico , Antígenos de Neoplasias/efectos de los fármacos , Antígenos de Neoplasias/metabolismo , Antineoplásicos Inmunológicos/uso terapéutico , Productos Biológicos/uso terapéutico , Supervivencia Celular/genética , Células Dendríticas/inmunología , Quimioterapia Combinada , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Herpesvirus Humano 1 , Humanos , Interferón Tipo I/metabolismo , Células Asesinas Naturales/inmunología , Células MCF-7 , Melanoma/patología , Virus Oncolíticos/genética , Simplexvirus/genética , Neoplasias Cutáneas/patología , Linfocitos T Citotóxicos/inmunología
5.
Proc Natl Acad Sci U S A ; 115(7): E1540-E1549, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29386395

RESUMEN

Inhibition of immune checkpoints programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) on T cells results in durable antitumor activity in melanoma patients. Despite high frequency of melanoma brain metastases (BrM) and associated poor prognosis, the activity and mechanisms of immune checkpoint inhibitors (ICI) in metastatic tumors that develop within the "immune specialized" brain microenvironment, remain elusive. We established a melanoma tumor transplantation model with intracranial plus extracranial (subcutaneous) tumor, mimicking the clinically observed coexistence of metastases inside and outside the brain. Strikingly, intracranial ICI efficacy was observed only when extracranial tumor was present. Extracranial tumor was also required for ICI-induced increase in CD8+ T cells, macrophages, and microglia in brain tumors, and for up-regulation of immune-regulatory genes. Combined PD-1/CTLA-4 blockade had a superior intracranial efficacy over the two monotherapies. Cell depletion studies revealed that NK cells and CD8+ T cells were required for intracranial anti-PD-1/anti-CTLA-4 efficacy. Rather than enhancing CD8+ T cell activation and expansion within intracranial tumors, PD-1/CTLA-4 blockade dramatically (∼14-fold) increased the trafficking of CD8+ T cells to the brain. This was mainly through the peripheral expansion of homing-competent effector CD8+ T cells and potentially further enhanced through up-regulation of T cell entry receptors intercellular adhesion molecule 1 and vascular adhesion molecule 1 on tumor vasculature. Our study indicates that extracranial activation/release of CD8+ T cells from PD-1/CTLA-4 inhibition and potentiation of their recruitment to the brain are paramount to the intracranial anti-PD-1/anti-CTLA-4 activity, suggesting augmentation of these processes as an immune therapy-enhancing strategy in metastatic brain cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/terapia , Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/terapia , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/secundario , Femenino , Granzimas/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/secundario , Neoplasias Cutáneas/terapia , Carga Tumoral , Células Tumorales Cultivadas
6.
Sci Transl Med ; 10(422)2018 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-29298869

RESUMEN

Immune checkpoint inhibitors, including those targeting programmed cell death protein 1 (PD-1), are reshaping cancer therapeutic strategies. Evidence suggests, however, that tumor response and patient survival are determined by tumor programmed death ligand 1 (PD-L1) expression. We hypothesized that preconditioning of the tumor immune microenvironment using targeted, virus-mediated interferon (IFN) stimulation would up-regulate tumor PD-L1 protein expression and increase cytotoxic T cell infiltration, improving the efficacy of subsequent checkpoint blockade. Oncolytic viruses (OVs) represent a promising form of cancer immunotherapy. For brain tumors, almost all studies to date have used direct intralesional injection of OV, because of the largely untested belief that intravenous administration will not deliver virus to this site. We show, in a window-of-opportunity clinical study, that intravenous infusion of oncolytic human Orthoreovirus (referred to herein as reovirus) leads to infection of tumor cells subsequently resected as part of standard clinical care, both in high-grade glioma and in brain metastases, and increases cytotoxic T cell tumor infiltration relative to patients not treated with virus. We further show that reovirus up-regulates IFN-regulated gene expression, as well as the PD-1/PD-L1 axis in tumors, via an IFN-mediated mechanism. Finally, we show that addition of PD-1 blockade to reovirus enhances systemic therapy in a preclinical glioma model. These results support the development of combined systemic immunovirotherapy strategies for the treatment of both primary and secondary tumors in the brain.


Asunto(s)
Neoplasias Encefálicas/terapia , Virus Oncolíticos/patogenicidad , Animales , Glioma/terapia , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/metabolismo
8.
J Nurs Manag ; 25(8): 624-631, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28857382

RESUMEN

AIM: To examine the nurse manager perspective surrounding implementation of unit level shared governance in one Veterans Health Administration facility. BACKGROUND: Nursing shared governance is a formal model allowing nursing staff decision-making input into clinical practice, quality improvement, evidence-based practice and staff professional development. Unit level shared governance is a management process where decision authority is delegated to nursing staff at the unit level. METHODS: Convenience sampling was used to recruit ten nurse managers who participated in face-to-face semi-structured interviews. Data were analysed using content analysis and constant comparison techniques. Demographic data were described using descriptive statistics. RESULTS: The participants included seven female and three male nurse managers with seven Caucasian and three African American. Participant quotes were clustered to identify sub-themes that were then grouped into four global themes to describe unit level shared governance. The global themes were: (1) motivation, (2) demotivation, (3) recommendations for success, and (4) outcomes. CONCLUSION: These research findings resonate with previous studies that shared governance may be associated with increased nurse empowerment, self-management, engagement, and satisfaction. IMPLICATIONS FOR NURSING MANAGEMENT: These findings reflect the need for nurse managers to promote and recognize staff participation in unit level shared governance.


Asunto(s)
Toma de Decisiones , Enfermeras Administradoras/psicología , Enfermeras y Enfermeros/normas , Percepción , Adulto , Femenino , Humanos , Masculino , Enfermeras y Enfermeros/psicología , Proyectos Piloto , Poder Psicológico , Investigación Cualitativa , Automanejo
9.
Mol Ther Oncolytics ; 5: 75-86, 2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28547002

RESUMEN

Pediatric high-grade glioma (pHGG) and diffuse intrinsic pontine glioma (DIPG) are invasive tumors with poor survival. Oncolytic virotherapy, initially devised as a direct cytotoxic treatment, is now also known to act via immune-mediated mechanisms. Here we investigate a previously unreported mechanism of action: the inhibition of migration and invasion in pediatric brain tumors. We evaluated the effect of oncolytic herpes simplex virus 1716 (HSV1716) on the migration and invasion of pHGG and DIPG both in vitro using 2D (scratch assay, live cell imaging) and 3D (spheroid invasion in collagen) assays and in vivo using an orthotopic xenograft model of DIPG invasion. HSV1716 inhibited migration and invasion in pHGG and DIPG cell lines. pHGG cells demonstrated reduced velocity and changed morphology in the presence of virus. HSV1716 altered pHGG cytoskeletal dynamics by stabilizing microtubules, inhibiting glycogen synthase kinase-3, and preventing localized clustering of adenomatous polyposis coli (APC) to the leading edge of cells. HSV1716 treatment also reduced tumor infiltration in a mouse orthotopic xenograft DIPG model. Our results demonstrate that HSV1716 targets the migration and invasion of pHGG and DIPG and indicates the potential of an oncolytic virus (OV) to be used as a novel anti-invasive treatment strategy for pediatric brain tumors.

10.
Viruses ; 7(12): 6291-312, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26633468

RESUMEN

The last few years have seen an increased interest in immunotherapy in the treatment of malignant disease. In particular, there has been significant enthusiasm for oncolytic virotherapy, with a large amount of pre-clinical data showing promise in animal models in a wide range of tumour types. How do we move forward into the clinical setting and translate something which has such potential into meaningful clinical outcomes? Here, we review how the field of oncolytic virotherapy has developed thus far and what the future may hold.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Viroterapia Oncolítica/tendencias
11.
Clin Cancer Res ; 19(18): 5104-15, 2013 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-23881927

RESUMEN

PURPOSE: Surgical removal of solid primary tumors is an essential component of cancer treatment. Surgery-induced dysfunction in natural killer (NK) cells has been linked to the development of metastases in animal models and patients with cancer. We investigated the activation of NK cells using influenza vaccine in the perioperative period to eradicate micrometastatic disease. EXPERIMENTAL DESIGN: Both the B16lacZ and 4T1 tumor models in immunocompetent mice were used to assess the in vivo efficacy of perioperative influenza vaccine administration. In healthy human donors and cancer surgery patients, we assessed NK cell function pre- and post-influenza vaccination using both in vivo and ex vivo assays. RESULTS: Using the TLR3 agonist poly(I:C), we showed as proof-of-principle that perioperative administration of a nonspecific innate immune stimulant can inhibit surgery-induced dysfunction in NK cells and attenuate metastases. Next, we assessed a panel of prophylactic vaccines for NK cell activation and determined that inactivated influenza vaccine was the best candidate for perioperative administration. Perioperative influenza vaccine significantly reduced tumor metastases and improved NK cytotoxicity in preclinical tumor models. Significantly, IFNα is the main cytokine mediator for the therapeutic effect of influenza vaccination. In human studies, influenza vaccine significantly enhanced NK cell activity in healthy human donors and cancer surgery patients. CONCLUSION: These results provide the preclinical rationale to pursue future clinical trials of perioperative NK cell activation, using vaccination in cancer surgery patients. Research into perioperative immune therapy is warranted to prevent immune dysfunction following surgery and eradicate metastatic disease.


Asunto(s)
Vacunas contra la Influenza/uso terapéutico , Células Asesinas Naturales/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/cirugía , Procedimientos Quirúrgicos Operativos/efectos adversos , Animales , Citotoxicidad Inmunológica/inmunología , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/cirugía , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Atención Perioperativa , Periodo Posoperatorio , Receptor de Interferón alfa y beta/fisiología , Células Tumorales Cultivadas , Vacunación
12.
Int J Cancer ; 132(10): 2327-38, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23114986

RESUMEN

Reovirus is a promising oncolytic virus, acting by both direct and immune-mediated mechanisms, although its potential may be limited by inactivation after systemic delivery. Our study addressed whether systemically delivered reovirus might be shielded from neutralising antibodies by cell carriage and whether virus-loaded blood or hepatic innate immune effector cells become activated to kill colorectal cancer cells metastatic to the liver in human systems. We found that reovirus was directly cytotoxic against tumour cells but not against fresh hepatocytes. Although direct tumour cell killing by neat virus was significantly reduced in the presence of neutralising serum, reovirus was protected when loaded onto peripheral blood mononuclear cells, which may carry virus after intravenous administration in patients. As well as handing off virus for direct oncolytic killing, natural killer (NK) cells within reovirus-treated blood mononuclear cells were stimulated to kill tumour targets, but not normal hepatocytes, in a Type I interferon-dependent manner. Similarly, NK cells within liver mononuclear cells became selectively cytotoxic towards tumour cells when activated by reovirus. Hence, intravenous reovirus may evade neutralisation by serum via binding to circulating mononuclear cells, and this blood cell carriage has the potential to investigate both direct and innate immune-mediated therapy against human colorectal or other cancers metastatic to the liver.


Asunto(s)
Adenocarcinoma/inmunología , Sangre/inmunología , Neoplasias del Colon/inmunología , Citotoxicidad Inmunológica , Leucocitos Mononucleares/inmunología , Neoplasias Hepáticas/inmunología , Hígado/citología , Viroterapia Oncolítica/métodos , Reoviridae/inmunología , Adenocarcinoma/patología , Línea Celular Tumoral , Neoplasias del Colon/patología , Neoplasias Colorrectales/inmunología , Citometría de Flujo , Hepatocitos/inmunología , Humanos , Infusiones Intravenosas , Células Asesinas Naturales/inmunología , Hígado/inmunología , Neoplasias Hepáticas/secundario , Fenotipo
13.
Sci Transl Med ; 4(138): 138ra77, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22700953

RESUMEN

Oncolytic viruses, which preferentially lyse cancer cells and stimulate an antitumor immune response, represent a promising approach to the treatment of cancer. However, how they evade the antiviral immune response and their selective delivery to, and replication in, tumor over normal tissue has not been investigated in humans. Here, we treated patients with a single cycle of intravenous reovirus before planned surgery to resect colorectal cancer metastases in the liver. Tracking the viral genome in the circulation showed that reovirus could be detected in plasma and blood mononuclear, granulocyte, and platelet cell compartments after infusion. Despite the presence of neutralizing antibodies before viral infusion in all patients, replication-competent reovirus that retained cytotoxicity was recovered from blood cells but not plasma, suggesting that transport by cells could protect virus for potential delivery to tumors. Analysis of surgical specimens demonstrated greater, preferential expression of reovirus protein in malignant cells compared to either tumor stroma or surrounding normal liver tissue. There was evidence of viral factories within tumor, and recovery of replicating virus from tumor (but not normal liver) was achieved in all four patients from whom fresh tissue was available. Hence, reovirus could be protected from neutralizing antibodies after systemic administration by immune cell carriage, which delivered reovirus to tumor. These findings suggest new preclinical and clinical scheduling and treatment combination strategies to enhance in vivo immune evasion and effective intravenous delivery of oncolytic viruses to patients in vivo.


Asunto(s)
Virus Oncolíticos/fisiología , Anciano , Anticuerpos Neutralizantes/inmunología , Plaquetas/virología , Neoplasias Colorrectales/cirugía , Neoplasias Colorrectales/terapia , Femenino , Genoma Viral/genética , Granulocitos/virología , Humanos , Neoplasias Hepáticas/cirugía , Neoplasias Hepáticas/terapia , Masculino , Persona de Mediana Edad , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Replicación Viral/genética , Replicación Viral/fisiología
14.
Biores Open Access ; 1(1): 3-15, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23515241

RESUMEN

Reovirus is a naturally occurring oncolytic virus that has shown preclinical efficacy in the treatment of a wide range of tumor types and has now reached phase III testing in clinical trials. The anti-cancer activity of reovirus has been attributed to both its direct oncolytic activity and the enhancement of anti-tumor immune responses. In this study, we have investigated the direct effect of reovirus on acute myeloid leukemia (AML) cells and its potential to enhance innate immune responses against AML, including the testing of primary samples from patients. Reovirus was found to replicate in and kill AML cell lines, and to reduce cell viability in primary AML samples. The pro-inflammatory cytokine interferon alpha (IFNα) and the chemokine (C-C motif) ligand 5 (known as RANTES [regulated upon activation, normal T-cell expressed, and secreted]) were also secreted from AML cells in response to virus treatment. In addition, reovirus-mediated activation of natural killer (NK) cells, within the context of peripheral blood mononuclear cells, stimulated their anti-leukemia response, with increased NK degranulation and IFNγ production and enhanced killing of AML targets. These data suggest that reovirus has the potential as both a direct cytotoxic and an immunotherapeutic agent for the treatment of AML.

15.
J Nurs Adm ; 39(12): 511-3, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19955962

RESUMEN

Using a systematic, evidence-based approach for developing a business plan allows nurse executives to forecast the needs of the organization, involve nursing staff at all levels, evaluate the direction of the profession, and present a plan with clear, concise goals. The authors describe 4 steps necessary in developing an effective evidence-based business plan.


Asunto(s)
Educación Continua en Enfermería/organización & administración , Enfermería Basada en la Evidencia/organización & administración , Enfermeras Administradoras/organización & administración , Servicio de Enfermería en Hospital/organización & administración , Desarrollo de Programa , Humanos , Liderazgo , Investigación en Administración de Enfermería , Innovación Organizacional , Técnicas de Planificación , Desarrollo de Personal/organización & administración , Estados Unidos
16.
BJU Int ; 103(6): 740-6, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19007376

RESUMEN

OBJECTIVE: To report the results of a programme aimed at determining the feasibility of autologous renal cell carcinoma (RCC) tissue collection and vaccine preparation within the setting of a UK National Health Service Cancer Centre. PATIENTS AND METHODS: Patients undergoing nephrectomy for suspected renal tumours were identified from theatre lists between April 2005 and July 2007. Samples of tumour were freshly cut from nephrectomy specimens. If tissue collection failed the reason was recorded prospectively. Cell viability was assessed after sample sieving. Freeze-thaw lysates were prepared from viable tumour cells, and the immunogenicity tested by pulsing onto dendritic cells (DC). RESULTS: Of 84 patients, 83 had a histological diagnosis of RCC; samples were obtained from 29 of these 83 (35%). Reasons for failure in tissue collection included that the tumour was too small or haemorrhagic/necrotic, pre-surgical embolization, and difficulties with fresh tumour collection out of normal working hours. Viable tumour cells were obtained in 12 of the 29 samples (41%); no factor was able to predict the production of viable cells. Unmodified lysates did not activate DC. CONCLUSION: An autologous RCC vaccination programme might fail to generate vaccines for a substantial proportion of eligible patients in the setting of a clinical cancer centre.


Asunto(s)
Vacunas contra el Cáncer , Carcinoma de Células Renales/prevención & control , Neoplasias Renales/prevención & control , Adyuvantes Inmunológicos , Adulto , Anciano , Anciano de 80 o más Años , Células Dendríticas/trasplante , Ensayo de Inmunoadsorción Enzimática , Métodos Epidemiológicos , Femenino , Citometría de Flujo , Humanos , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Masculino , Persona de Mediana Edad , Nefrectomía , Evaluación de Programas y Proyectos de Salud , Bancos de Tejidos
17.
Clin Cancer Res ; 14(22): 7358-66, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19010851

RESUMEN

PURPOSE: Early clinical trials are under way exploring the direct oncolytic potential of reovirus. This study addresses whether tumor infection by reovirus is also able to generate bystander, adaptive antitumor immunity. EXPERIMENTAL DESIGN: Reovirus was delivered intravenously to C57BL/6 mice bearing lymph node metastases from the murine melanoma, B16-tk, with assessment of nodal metastatic clearance, priming of antitumor immunity against the tumor-associated antigen tyrosinase-related protein-2, and cytokine responses. In an in vitro human system, the effect of reovirus infection on the ability of Mel888 melanoma cells to activate and load dendritic cells for cytotoxic lymphocyte (CTL) priming was investigated. RESULTS: In the murine model, a single intravenous dose of reovirus reduced metastatic lymph node burden and induced antitumor immunity (splenocyte response to tyrosinase-related protein-2 and interleukin-12 production in disaggregated lymph nodes). In vitro human assays revealed that uninfected Mel888 cells failed to induce dendritic cell maturation or support priming of an anti-Mel888 CTL response. In contrast, reovirus-infected Mel888 cells (reo-Mel) matured dendritic cells in a reovirus dose-dependent manner. When cultured with autologous peripheral blood lymphocytes, dendritic cells loaded with reo-Mel induced lymphocyte expansion, IFN-gamma production, specific anti-Mel888 cell cytotoxicity, and cross-primed CD8+ T cells specific against the human tumor-associated antigen MART-1. CONCLUSION: Reovirus infection of tumor cells reduces metastatic disease burden and primes antitumor immunity. Future clinical trials should be designed to explore both direct cytotoxic and immunotherapeutic effects of reovirus.


Asunto(s)
Antineoplásicos/uso terapéutico , Citotoxicidad Inmunológica/inmunología , Neoplasias Experimentales/terapia , Viroterapia Oncolítica/métodos , Infecciones por Reoviridae/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interferón gamma/biosíntesis , Oxidorreductasas Intramoleculares/inmunología , Metástasis Linfática/patología , Activación de Linfocitos/inmunología , Antígeno MART-1 , Orthoreovirus Mamífero 3/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/virología , Reacción en Cadena de la Polimerasa , Linfocitos T Citotóxicos/inmunología
18.
Environ Sci Technol ; 36(6): 1245-56, 2002 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11944676

RESUMEN

Gaseous elemental mercury (Hg0) is a globally distributed air toxin with a long atmospheric residence time. Any process that reduces its atmospheric lifetime increases its potential accumulation in the biosphere. Our data from Barrow, AK, at 71 degrees N show that rapid, photochemically driven oxidation of boundary-layer Hg0 after polar sunrise, probably by reactive halogens, creates a rapidly depositing species of oxidized gaseous mercury in the remote Arctic troposphere at concentrations in excess of 900 pg m(-3). This mercury accumulates in the snowpack during polar spring at an accelerated rate in a form that is bioavailable to bacteria and is released with snowmelt during the summer emergence of the Arctic ecosystem. Evidence suggests that this is a recent phenomenon that may be occurring throughout the earth's polar regions.


Asunto(s)
Contaminantes Atmosféricos/análisis , Mercurio/química , Regiones Árticas , Bacterias , Disponibilidad Biológica , Ecosistema , Monitoreo del Ambiente , Gases , Mercurio/análisis , Oxidación-Reducción , Periodicidad , Fotoquímica , Nieve
19.
Environ Sci Technol ; 36(23): 5034-40, 2002 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-12523417

RESUMEN

The METAALICUS (Mercury Experiment To Assess Atmospheric Loading In Canada and the US) project is a whole ecosystem experiment designed to study the activity, mobility, and availability of atmospherically deposited mercury. To investigate the dynamics of mercury newly deposited onto a terrestrial ecosystem, an enriched stable isotope of mercury (202Hg) was sprayed onto a Boreal forest subcatchment in an experiment that allowed us, for the first time, to monitor the fate of 'new' mercury in deposition and to distinguish it from native mercury historically stored in the ecosystem. Newly deposited mercury was more reactive than the native mercury with respect to volatilization and methylation pathways. Mobility through runoff was very low and strongly decreased with time because of a rapid equilibration with the large native pool of "bound" mercury. Over one season, only approximately 8% of the added 212Hg volatilized to the atmosphere and less than 1% appeared in runoff. Within a few months, approximately 66% of the applied 202Hg remained associated with above ground vegetation, with the rest being incorporated into soils. The fraction of 202Hg bound to vegetation was much higher than seen for native Hg (<5% vegetation), suggesting that atmospherically derived mercury enters the soil pool with a time delay, after plants senesce and decompose. The initial mobility of mercury received through small rain events or dry deposition decreased markedly in a relatively short time period, suggesting that mercury levels in terrestrial runoff may respond slowly to changes in mercury deposition rates.


Asunto(s)
Ecosistema , Mercurio/análisis , Árboles , Adsorción , Biodegradación Ambiental , Disponibilidad Biológica , Canadá , Monitoreo del Ambiente , Mercurio/química , Plantas , Estaciones del Año , Estados Unidos , Movimientos del Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...