Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Langenbecks Arch Surg ; 406(3): 597-605, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33301071

RESUMEN

PURPOSE: The reported conversion rates for minimally invasive distal pancreatectomy (MIDP) range widely from 2 to 38%. The identification of risk factors for conversion may help surgeons during preoperative planning and patient counseling. Moreover, the impact of conversion on outcomes of MIDP is unknown. METHODS: A systematic review was conducted as part of the 2019 Miami International Evidence-Based Guidelines on Minimally Invasive Pancreas Resection (IG-MIPR). The PubMed, Cochrane, and Embase databases were searched for studies concerning conversion to open surgery in MIDP. RESULTS: Of the 828 studies screened, eight met the eligibility criteria, resulting in a combined dataset including 2592 patients after MIDP. The overall conversion rate was 17.1% (range 13.0-32.7%) with heterogeneity between studies associated with the definition of conversion adopted. Only one study divided conversion into elective and emergency conversion. The main indications for conversion were vascular involvement (23.7%), concern for oncological radicality (21.9%), and bleeding (18.9%). The reported risk factors for conversion included a malignancy as an indication for surgery, the proximity of the tumor to vascular structures in preoperative imaging, higher BMI or visceral fat, and multi-organ resection or extended resection. Contrasting results were seen in terms of blood loss and length of stay in comparing converted MIDP and completed MIDP patients. CONCLUSION: The identified risk factors for conversion from this study can be used for patient selection and counseling. Surgeon experience should be considered when contemplating MIDP for a complex patient. Future studies should divide conversion into elective and emergency conversion.


Asunto(s)
Laparoscopía , Neoplasias Pancreáticas , Procedimientos Quirúrgicos Robotizados , Humanos , Pancreatectomía , Neoplasias Pancreáticas/cirugía , Factores de Riesgo , Resultado del Tratamiento
2.
Eur J Surg Oncol ; 44(9): 1378-1383, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30131104

RESUMEN

BACKGROUND: Peritoneal metastasis from biliary carcinoma (PMC) is associated with poor prognosis when treated with chemotherapy. OBJECTIVE: To evaluate the impact on survival of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), and compare with conventional palliative chemotherapy for patients with PMC. MATERIAL AND METHODS: A prospective multicenter international database was retrospectively searched to identify all patients with PMC treated with a potentially curative CRS/HIPEC (CRS/HIPEC group). The overall survival (OS) was compared to patients with PMC treated with palliative chemotherapy (systemic chemotherapy group). Survival was analyzed using Kaplan-Meier method and compared with Log-Rank test. RESULTS: Between 1995 and 2015, 34 patients were included in the surgical group, and compared to 21 in the systemic chemotherapy group. In the surgical group, median peritoneal cancer index was 9 (range 3-26), macroscopically complete resection was obtained for 25 patients (73%). There was more gallbladder localization in the surgical group compared to the chemotherapy group (35% vs. 18%, p = 0.001). Median OS was 21.4 and 9.3 months for surgical and chemotherapy group, respectively (p=0.007). Three-year overall survival was 30% and 10% for surgical and chemotherapy group, respectively. CONCLUSION: Treatment with CRS and HIPEC for biliary carcinoma with peritoneal metastasis is feasible and may provide survival benefit when compared to palliative chemotherapy.


Asunto(s)
Neoplasias de los Conductos Biliares/terapia , Procedimientos Quirúrgicos de Citorreducción/métodos , Hipertermia Inducida/métodos , Neoplasias Peritoneales/terapia , Sistema de Registros , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de los Conductos Biliares/mortalidad , Neoplasias de los Conductos Biliares/secundario , Femenino , Estudios de Seguimiento , Francia/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Peritoneales/mortalidad , Neoplasias Peritoneales/secundario , Pronóstico , Estudios Prospectivos , Tasa de Supervivencia/tendencias
3.
J Pancreat Cancer ; 3(1): 1-4, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29082362

RESUMEN

BACKGROUND: Pancreatic panniculitis is a rare cause of subcutaneous fat necrosis secondary to elevated serum levels of pancreatic enzymes. It is most often associated with pancreatic acinar cell carcinoma, but has also been seen in patients with pancreatitis. CASE REPORT: We present a case of a 64 year old Caucasian man without symptoms of pancreatitis who presents with pancreatic panniculitis manifesting in multiple subcutaneous ulcerating nodules of the bilateral lower extremities, discovered to have a previously unreported etiology for this condition. He had no evidence of pancreatitis or malignancy, but instead a pancreatic-portal fistula resulting in panniculitis. CONCLUSION: Peripancreatic vascular lesions must also be considered in the differential diagnosis of pancreatic panniculitis. The diagnosis, pathology, and treatment of pancreatic panniculitis are reviewed herein.

4.
Cancer Gene Ther ; 22(6): 326-34, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25908451

RESUMEN

Neutrophil extracellular traps (NETs) are formed when neutrophils expel their DNA, histones and intracellular proteins into the extracellular space or circulation. NET formation is dependent on autophagy and is mediated by citrullination of histones to allow for the unwinding and subsequent expulsion of DNA. NETs have an important role in the pathogenesis of several sterile inflammatory diseases, including malignancy, therefore we investigated the role of NETs in the setting of pancreatic ductal adenocarcinoma (PDA). Neutrophils isolated from two distinct animal models of PDA had an increased propensity to form NETs following stimulation with platelet activating factor (PAF). Serum DNA, a marker of circulating NET formation, was elevated in tumor bearing animals as well as in patients with PDA. Citrullinated histone H3 expression, a marker of NET formation, was observed in pancreatic tumors obtained from murine models and patients with PDA. Inhibition of autophagy with chloroquine or genetic ablation of receptor for advanced glycation end products (RAGE) resulted in decreased propensity for NET formation, decreased serum DNA and decreased citrullinated histone H3 expression in the pancreatic tumor microenvironment. We conclude that NETs are upregulated in pancreatic cancer through RAGE-dependent/autophagy mediated pathways.


Asunto(s)
Autofagia , Carcinoma Ductal Pancreático/fisiopatología , Trampas Extracelulares/fisiología , Neutrófilos/fisiología , Neoplasias Pancreáticas/fisiopatología , Receptor para Productos Finales de Glicación Avanzada/fisiología , Animales , Carcinoma Ductal Pancreático/inmunología , Femenino , Humanos , Ratones , Ratones Noqueados , Neoplasias Pancreáticas/inmunología , Receptor para Productos Finales de Glicación Avanzada/genética
5.
Cell Death Dis ; 5: e1480, 2014 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-25341034

RESUMEN

A hypoxic tumor microenvironment is characteristic of many cancer types, including one of the most lethal, pancreatic cancer. We recently demonstrated that the receptor for advanced glycation end products (RAGE) has an important role in promoting the development of pancreatic cancer and attenuating the response to chemotherapy. We now demonstrate that binding of RAGE to oncogenic KRAS facilitates hypoxia-inducible factor 1 (HIF1)α activation and promotes pancreatic tumor growth under hypoxic conditions. Hypoxia induces NF-κB-dependent and HIF1α-independent RAGE expression in pancreatic tumor cells. Moreover, the interaction between RAGE and mutant KRAS increases under hypoxia, which in turn sustains KRAS signaling pathways (RAF-MEK-ERK and PI3K-AKT), facilitating stabilization and transcriptional activity of HIF1α. Knock down of RAGE in vitro inhibits KRAS signaling, promotes HIF1α degradation, and increases hypoxia-induced pancreatic tumor cell death. RAGE-deficient mice have impaired oncogenic KRAS-driven pancreatic tumor growth with significant downregulation of the HIF1α signaling pathway. Our results provide a novel mechanistic link between NF-κB, KRAS, and HIF1α, three potent molecular pathways in the cellular response to hypoxia during pancreatic tumor development and suggest alternatives for preventive and therapeutic strategies.


Asunto(s)
Oncogenes , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Animales , Autofagia , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , FN-kappa B/metabolismo , Estabilidad Proteica , Receptor para Productos Finales de Glicación Avanzada , Activación Transcripcional
6.
Oncogene ; 33(5): 567-77, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-23318458

RESUMEN

Tumor cells require increased adenosine triphosphate (ATP) to support anabolism and proliferation. The precise mechanisms regulating this process in tumor cells are unknown. Here, we show that the receptor for advanced glycation endproducts (RAGE) and one of its primary ligands, high-mobility group box 1 (HMGB1), are required for optimal mitochondrial function within tumors. We found that RAGE is present in the mitochondria of cultured tumor cells as well as primary tumors. RAGE and HMGB1 coordinately enhanced tumor cell mitochondrial complex I activity, ATP production, tumor cell proliferation and migration. Lack of RAGE or inhibition of HMGB1 release diminished ATP production and slowed tumor growth in vitro and in vivo. These findings link, for the first time, the HMGB1-RAGE pathway with changes in bioenergetics. Moreover, our observations provide a novel mechanism within the tumor microenvironment by which necrosis and inflammation promote tumor progression.


Asunto(s)
Complejo I de Transporte de Electrón/metabolismo , Proteína HMGB1/metabolismo , Neoplasias Pancreáticas/patología , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Adenosina Trifosfato/biosíntesis , Adenosina Trifosfato/metabolismo , Animales , Butadienos/farmacología , Antígeno CD24/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Cicloheximida/farmacología , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Metabolismo Energético , Inhibidores Enzimáticos/farmacología , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteína HMGB1/efectos de los fármacos , Humanos , Inflamación/metabolismo , MAP Quinasa Quinasa 2/genética , MAP Quinasa Quinasa 2/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Nitrilos/farmacología , Neoplasias Pancreáticas/metabolismo , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Inhibidores de la Síntesis de la Proteína/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor para Productos Finales de Glicación Avanzada/genética , Rotenona/farmacología , Transducción de Señal , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Microambiente Tumoral , Desacopladores
7.
Ann Surg Oncol ; 20(7): 2180-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23575907

RESUMEN

BACKGROUND: Isolated hepatic perfusion (IHP) with melphalan is an established approach for patients with unresectable metastatic liver lesions. This study determined the safety and maximum tolerated dose (MTD) of 5-FU with oxaliplatin via IHP. METHODS: Standard 3 × 3 Phase I design. Subjects with unresectable isolated CRC liver metastases scheduled for HAI pump were eligible. IHP used fixed-dose oxaliplatin with escalating 5-FU doses. Toxicity (CTCAE v 4.0) and response (RECIST), progression-free survival, and overall survival (OS) were assessed. Systemic and IHP plasma PK of 5-FU, anabolites, and platinum were determined. RESULTS: All 12 patients had received ≥ 1 line of pre-IHP chemotherapy. There were 4 grade 3 serious adverse events (33.3 %) and 1 grade 4 event (8.3 %). Also, 2 dose-limiting toxicities occurred at DL2 at 300 mg/m(2), resulting in expansion of DL1 at 200 mg/m(2) 5-FU, the eventual MTD. At 6-month follow-up, 9 patients (82 %) demonstrated partial response, while 2 (18 %) exhibited stable disease. Also, 64 % of patients demonstrated a >50 % decrease in CEA. The 1- and 2-year OS probabilities were 90.9 and 71.6 %, respectively, with median follow-up of 24 months. IHP exposures (AUC0-60 min) were 10.9 ± 4.5 µgPt h/mL, 49.3 ± 30.7 µg h/mL 5-FU (DL1), and 70.5 ± 35.5 µg h/mL 5-FU (DL2). Systemic exposure (AUC0-inf) relative to IHP exposure was negligible for both platinum (1.1 ± 1.5 %) and 5-FU (0.09 ± 0.10 %). CONCLUSIONS: The MTD for IHP was 200 mg/m(2) 5-FU with 40 mg/m(2) oxaliplatin. Systemic exposure to the agents was minimal during IHP. The response and survival observed warrants assessment in a larger phase II trial.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Quimioterapia del Cáncer por Perfusión Regional , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/tratamiento farmacológico , Dosis Máxima Tolerada , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Área Bajo la Curva , Antígeno Carcinoembrionario/sangre , Quimioterapia del Cáncer por Perfusión Regional/efectos adversos , Neoplasias Colorrectales/sangre , Supervivencia sin Enfermedad , Femenino , Fluorouracilo/administración & dosificación , Fluorouracilo/farmacocinética , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/sangre , Neoplasias Hepáticas/secundario , Masculino , Persona de Mediana Edad , Compuestos Organoplatinos/administración & dosificación , Compuestos Organoplatinos/farmacocinética , Oxaliplatino
8.
Adv Surg ; 45: 323-40, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21954697

RESUMEN

Robotic-assisted major pancreatic resections allow recreation of time-tested open surgical procedures on a minimally invasive platform. Early outcomes from robotic-assisted major pancreatic resections are comparable with those of laparoscopic and open approaches. Robotic assistance has the potential to bring the well-recognized advantages of minimally invasive surgery to major pancreatic resections. Technological innovations and increased surgeon familiarity with this approach will improve, likely leading to greater adoption and acceptance.


Asunto(s)
Pancreatectomía/métodos , Enfermedades Pancreáticas/cirugía , Pancreaticoduodenectomía/métodos , Robótica , Adenocarcinoma/cirugía , Disección , Humanos , Laparoscopía , Tumores Neuroendocrinos/cirugía , Neoplasias Pancreáticas/cirugía , Vena Porta/cirugía , Robótica/métodos , Resultado del Tratamiento
9.
Cell Death Differ ; 18(4): 571-80, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21311563

RESUMEN

Beclin 1, the mammalian orthologue of yeast Atg6, has a central role in autophagy, a process of programmed cell survival, which is increased during periods of cell stress and extinguished during the cell cycle. It interacts with several cofactors (Atg14L, UVRAG, Bif-1, Rubicon, Ambra1, HMGB1, nPIST, VMP1, SLAM, IP(3)R, PINK and survivin) to regulate the lipid kinase Vps-34 protein and promote formation of Beclin 1-Vps34-Vps15 core complexes, thereby inducing autophagy. In contrast, the BH3 domain of Beclin 1 is bound to, and inhibited by Bcl-2 or Bcl-XL. This interaction can be disrupted by phosphorylation of Bcl-2 and Beclin 1, or ubiquitination of Beclin 1. Interestingly, caspase-mediated cleavage of Beclin 1 promotes crosstalk between apoptosis and autophagy. Beclin 1 dysfunction has been implicated in many disorders, including cancer and neurodegeneration. Here, we summarize new findings regarding the organization and function of the Beclin 1 network in cellular homeostasis, focusing on the cross-regulation between apoptosis and autophagy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis , Autofagia , Proteínas de la Membrana/fisiología , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Fosfatidilinositol 3-Quinasas Clase III/metabolismo , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína de Clasificación Vacuolar VPS15/metabolismo , Proteína bcl-X/metabolismo
10.
Oncogene ; 30(16): 1851-4, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21151175

RESUMEN

One hallmark of cancer is intrinsic or acquired resistance to apoptosis. Surprisingly, recent studies demonstrate that CD95/Fas/Apo1 and p53 upregulated mediator of apoptosis/PUMA (potent inducers of the death receptor and the mitochondrial apoptotic pathways, respectively) promote tumorigenesis. These findings provide important insights into the multifaceted roles of apoptosis in tumorigenesis.


Asunto(s)
Apoptosis , Transformación Celular Neoplásica , Humanos
11.
Oncogene ; 29(38): 5299-310, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20622903

RESUMEN

The functional relationship and cross-regulation between autophagy and apoptosis is complex. In this study we show that the high-mobility group box 1 protein (HMGB1) is a redox-sensitive regulator of the balance between autophagy and apoptosis. In cancer cells, anticancer agents enhanced autophagy and apoptosis, as well as HMGB1 release. HMGB1 release may be a prosurvival signal for residual cells after various cytotoxic cancer treatments. Diminished HMGB1 by short hairpin RNA transfection or inhibition of HMGB1 release by ethyl pyruvate or other small molecules led predominantly to apoptosis and decreased autophagy in stressed cancer cells. In this setting, reducible HMGB1 binds to the receptor for advanced glycation end products (RAGEs), but not to Toll-like receptor 4, induces Beclin1-dependent autophagy and promotes tumor resistance to alkylators (melphalan), tubulin disrupting agents (paclitaxel), DNA crosslinkers (ultraviolet light) and DNA intercalators (oxaliplatin or adriamycin). On the contrary, oxidized HMGB1 increases the cytotoxicity of these agents and induces apoptosis mediated by the caspase-9/-3 intrinsic pathway. HMGB1 release, as well as its redox state, thus links autophagy and apoptosis, representing a suitable target when coupled with conventional tumor treatments.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Proteína HMGB1/fisiología , Neoplasias/patología , Antineoplásicos/farmacología , Proteína HMGB1/metabolismo , Oxidación-Reducción
12.
Cell Death Differ ; 17(4): 666-76, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19834494

RESUMEN

Activation of the induced receptor for advanced glycation end products (RAGE) leads to initiation of NF-kappaB and MAP kinase signaling pathways, resulting in propagation and perpetuation of inflammation. RAGE-knockout animals are less susceptible to acute inflammation and carcinogen-induced tumor development. We have reported that most forms of tumor cell death result in release of the RAGE ligand, high-mobility group protein 1 (HMGB1). We now report a novel role for RAGE in the tumor cell response to stress. Targeted knockdown of RAGE in the tumor cell, leads to increased apoptosis, diminished autophagy and decreased tumor cell survival . In contrast, overexpression of RAGE is associated with enhanced autophagy, diminished apoptosis and greater tumor cell viability. RAGE limits apoptosis through a p53-dependent mitochondrial pathway. Moreover, RAGE-sustained autophagy is associated with decreased phosphorylation of mammalian target of rapamycin (mTOR) and increased Beclin-1/VPS34 autophagosome formation. These findings show that the inflammatory receptor, RAGE, has a heretofore unrecognized role in the tumor cell response to stress. Furthermore, these studies establish a direct link between inflammatory mediators in the tumor microenvironment and resistance to programmed cell death. Our data suggest that targeted inhibition of RAGE or its ligands may serve as novel targets to enhance current cancer therapies.


Asunto(s)
Apoptosis/fisiología , Autofagia/fisiología , Carcinoma/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores Inmunológicos/metabolismo , Estrés Fisiológico/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Carcinoma/fisiopatología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Proteína HMGB1/metabolismo , Humanos , Inflamación/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Pancreáticas/fisiopatología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/genética , Serina-Treonina Quinasas TOR , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
13.
Cancer Gene Ther ; 15(2): 115-25, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18084242

RESUMEN

In this study, we assessed the ability of a highly tumor-selective oncolytic vaccinia virus armed with a yeast cytosine deaminase gene to infect and lyse human and murine ovarian tumors both in vitro and in vivo. The virus vvDD-CD could infect, replicate in and effectively lyse both human and mouse ovarian cancer cells in vitro. In two different treatment schedules involving either murine MOSEC or human A2780 ovarian carcinomatosis models, regional delivery of vvDD-CD selectively targeted tumor cells and ovarian tissue, effectively delaying the development of either tumor or ascites and leading to significant survival advantages. Oncolytic virotherapy using vvDD-CD in combination with the prodrug 5-fluorocytosine conferred an additional long-term survival advantage upon tumor-bearing immunocompetent mice. These findings demonstrate that a tumor-selective oncolytic vaccinia combined with gene-directed enzyme prodrug therapy is a highly effective strategy for treating advanced ovarian cancers in both syngeneic mouse and human xenograft models. Given the biological safety, tumor selectivity and oncolytic potency of this armed oncolytic virus, this dual therapy merits further investigation as a promising new treatment for metastatic ovarian cancer.


Asunto(s)
Carcinoma/terapia , Citosina Desaminasa/genética , Viroterapia Oncolítica , Neoplasias Ováricas/terapia , Saccharomyces cerevisiae/genética , Virus Vaccinia/genética , Replicación Viral , Animales , Antimetabolitos/administración & dosificación , Antimetabolitos/uso terapéutico , Carcinoma/tratamiento farmacológico , Línea Celular Tumoral , Terapia Combinada , Citosina Desaminasa/administración & dosificación , Citosina Desaminasa/uso terapéutico , Femenino , Flucitosina/administración & dosificación , Flucitosina/uso terapéutico , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Neoplasias Ováricas/tratamiento farmacológico , Saccharomyces cerevisiae/enzimología , Virus Vaccinia/fisiología , Replicación Viral/genética
14.
Gene Ther ; 14(8): 638-47, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17268533

RESUMEN

To enhance further the safety and efficacy of oncolytic vaccinia virus, we have developed a new virus with targeted deletions of three viral genes encoding thymidine kinase and antiapoptotic/host range proteins SPI-1 and SPI-2 (vSPT). Infection of human and murine tumor cell lines yielded nearly equivalent or a log lower virus recovery in comparison to parental viruses. Viral infection activated multiple caspases in cancer cells but not in normal cells, suggesting infected cells may die via different pathways. In tumor-bearing mice, vSPT recovery from MC38 tumor was slightly reduced in comparison to two parental viruses. However, no virus was recovered from the brains and livers of mice injected with vSPT in contrast to control viruses. vSPT demonstrated significantly lower pathogenicity in nude mice. Systemic delivery of vSPT showed significant tumor inhibition in subcutaneous MC38 tumor, human ovarian A2780 and murine ovarian MOSEC carcinomatosis models; however, the tumor inhibition by vSPT was reduced compared with parental viruses. These results demonstrated that although deletion of these three viral genes further enhanced tumor selectivity, it also weakened the oncolytic potency. This study illustrates the complexity of creating a tumor-selective oncolytic virus by deleting multiple viral genes involved in multiple cellular pathways.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Virus Vaccinia/genética , Animales , Línea Celular Tumoral , Femenino , Eliminación de Gen , Marcación de Gen/métodos , Ingeniería Genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Modelos Animales , Trasplante de Neoplasias , Proteínas Quinasas/genética , Proteínas Recombinantes de Fusión/genética , Seguridad , Factor de Transcripción Sp2/genética , Timidina Quinasa/genética , Trasplante Heterólogo , Replicación Viral
15.
Cancer Biomark ; 1(6): 259-69, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-17192050

RESUMEN

Early detection of pancreatic cancer might improve clinical outcome. Significant alterations in the levels of individual serum cytokines have been reported in pancreatic cancer. We hypothesized that a multicytokine panel could serve as biomarkers for pancreatic cancer. To evaluate the diagnostic utility of such a panel, we have utilized a novel multianalyte LabMAP profiling technology that allows simultaneous measurement of multiple markers. In this study, a panel of 31 serological markers including cytokines, chemokines, growth and angiogenic factors in combination with CA 19-9 was analyzed in sera of pancreatic cancer patients, patients with chronic pancreatitis, and matched control healthy subjects. Statistical analysis identified a multicytokine panel that was able to distinguish pancreatic cancer from healthy controls with a sensitivity of 85.7% and specificity of 92.3%, which was superior to performance of CA 19-9 alone. Importantly, a multicytokine panel allowed the discrimination of pancreatic cancer from chronic pancreatitis with high sensitivity of 98% and specificity of 96.4%. In conclusion, we demonstrated that analysis of multiple serum cytokines using a novel LabMAP technology is a promising approach for development of a diagnostic assay for pancreatic cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Citocinas/sangre , Neoplasias Pancreáticas/diagnóstico , Análisis por Matrices de Proteínas/métodos , Antígeno CA-19-9/sangre , Estudios de Casos y Controles , Grupos Control , Interpretación Estadística de Datos , Diagnóstico Diferencial , Femenino , Humanos , Masculino , Neoplasias Pancreáticas/sangre , Pancreatitis Crónica/sangre , Pancreatitis Crónica/diagnóstico , Sensibilidad y Especificidad
16.
Trends Mol Med ; 7(7): 307-13, 2001 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-11425639

RESUMEN

Despite recent advances in the treatment of colorectal cancer, the overall survival rate for those patients with advanced locoregional disease remains less than 50%. Although adjuvant systemic chemotherapy has improved survival of these patients, more effective therapies are needed. Immunotherapy is an approach that could have a particular role in the adjuvant therapy of colorectal cancer. There is now convincing evidence that the immune system can specifically recognize and destroy malignant cells. Although both antibody- and T-cell-mediated anti-tumor responses have been documented, the cellular immune response with its direct cytotoxic mechanisms is felt to be the principal anti-tumor arm of the immune system. Analysis of the T cells that recognize tumors has led to the identification and characterization of many tumor-associated antigens including several colorectal antigens. Current approaches to developing a vaccine for colorectal cancer use our expanded understanding of these tumor-associated antigens and the conditions that allow development of an effective cellular immune response to them.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Vacuna BCG , Células Dendríticas/inmunología , Humanos , Inmunoterapia/métodos , Vacunas Sintéticas
17.
J Immunol ; 162(2): 989-94, 1999 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-9916724

RESUMEN

A majority of the human tumor-associated Ags characterized to date are derived from nonmutated "self"-proteins. Little is currently understood about the nature of the self-reactive lymphocytes that recognize these Ags. We recently characterized two nonmutated tumor-associated Ags for the B16 murine melanoma: tyrosinase-related protein-2 (TRP-2) and the endogenous retroviral envelope protein, p15E. We previously reported that both TRP-2 and p15E reactive CTL could be detected in the spleens of naive animals after a single in vitro stimulation using 10(-5)-10(-6) M of the appropriate Kb-binding 9-amino acid epitope. In this report we show that the CTL found in naive animals are low avidity lymphocytes, that respond only to high concentrations of peptide in vitro. We demonstrate that titration of in vitro-stimulating peptide to limiting concentrations distinguishes qualitative differences in the lymphocyte reactivity to these two Ags between vaccinated and unvaccinated animals. We further demonstrate that in vitro expansion of CTL in either high or low concentrations of stimulating peptide generated CTL cultures with different avidities for the relevant epitopes. CTL expanded in low concentrations demonstrated higher avidity for peptide-pulsed targets and better tumor recognition, when compared to CTL generated in the presence of high concentrations of Ag. More importantly, high avidity CTL demonstrated superior in vivo antitumor activity. These results demonstrate that qualitative differences in the CTL that recognize these two self-Ags are critically important to their in vitro and in vivo anti-tumor efficacy.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/uso terapéutico , Citotoxicidad Inmunológica , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular , Células Cultivadas , Pruebas Inmunológicas de Citotoxicidad , Relación Dosis-Respuesta Inmunológica , Femenino , Inyecciones Intravenosas , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Oligopéptidos/administración & dosificación , Oligopéptidos/inmunología , Oligopéptidos/uso terapéutico , Linfocitos T Citotóxicos/metabolismo , Células Tumorales Cultivadas
18.
J Immunol ; 154(12): 6466-74, 1995 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-7759882

RESUMEN

Our recent studies using IL-12 protein or fibroblasts genetically engineered to secrete IL-12 have demonstrated profound antitumor effects of IL-12 in murine models. The antitumor effects of local, high level IL-12 expression were examined using a retroviral vector, which can express both IL-12 subunits (p35 and p40) and the neomycin phosphotransferase (Neo)-marker gene from a polycistronic message utilizing internal ribosome entry site sequences. All animals intradermally (i.d.) receiving MCA207 murine sarcoma cell line nontransfected or Neo-transfected had progressively growing tumor, whereas all animals injected with MCA207 transfected with IL-12 were tumor free and were subsequently determined to be immune to a rechallenge of nontransfected MCA207 i.d. Similar results were obtained in experiments using the poorly immunogenic MCA102 murine sarcoma cell line. The inoculation of live MCA207-IL-12 tumor cells also caused the regression of contralateral nontransfected MCA207 inoculated either at the same time (80% protection) or up to 3 days before (33% protection) to the therapeutic tumor inoculation. In vivo depletion studies suggest that NK cells and IFN-gamma play important roles in the development of the early phase of the antitumor response, but that T cells (both CD4+ and CD8+) play the major role in the subsequent events, leading to long-term immunity. The potent antitumor effects observed for paracrine gene-delivered administration of IL-12 have thus been confirmed for multiple tumor cell types and in multiple murine strains. We believe that these results support the feasibility of IL-12 gene therapy for the treatment of human cancer.


Asunto(s)
Terapia Genética , Interleucina-12/genética , Neoplasias Experimentales/terapia , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Expresión Génica , Vectores Genéticos , Interleucina-12/antagonistas & inhibidores , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Pruebas de Neutralización , Retroviridae/genética , Sarcoma Experimental/inmunología , Sarcoma Experimental/terapia , Transfección , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
19.
Hum Immunol ; 39(2): 79-86, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8175386

RESUMEN

A novel class I-peptide-binding assay was developed and used to identify a series of peptides derived from the human p53 tumor-suppressor gene product capable of binding the HLA-A2 class I allele. Brief pH 3.3 acid treatment of human cell lines rapidly denatures pre-existing class I complexes, as detected by loss of binding of conformation-dependent mAbs, leaving only free class I heavy chains associated with the viable cell surface. These heavy chains may be induced to refold and be recognized by antibodies (in 2-4 hours) when acid-treated cells are coincubated with exogenous beta 2-microglobulin and peptides capable of binding the relevant class I allele examined. This assay, with a detection limit of 1-10 nM peptide, was used to screen the capacity of a panel of nine peptides bearing HLA-A2-binding motifs and derived from the human p53 tumor-suppressor protein sequence. Eight of the nine peptides bound to, and reconstituted, HLA-A2 on acid-treated cells. This assay system will enable the rapid identification of peptides binding to any class I allele, which is the initial prerequisite for elucidating potential CD8+ T-cell epitopes.


Asunto(s)
Antígeno HLA-A2/análisis , Linfocitos T/inmunología , Proteína p53 Supresora de Tumor/inmunología , Secuencia de Aminoácidos , Línea Celular , Citometría de Flujo , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Proteína p53 Supresora de Tumor/análisis
20.
Cancer Res ; 54(1): 182-9, 1994 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-7903204

RESUMEN

Interleukin 12 (IL-12), a disulfide-linked heterodimeric cytokine produced primarily by macrophages, is composed of light (p35) and heavy (p40) chains. It binds to a receptor on T-cells and natural killer cells, promoting the induction of primarily a TH1 response in vitro and in vivo. To determine whether paracrine IL-12 secretion can alter tumor cell growth or promote antitumor immunity, we have developed a delivery system using genetically engineered fibroblasts in murine tumor models. NIH3T3 cells were stably transfected to express 100-240 units/10(6) cells/48 h of IL-12 using expression plasmids carrying both the murine p35 and p40 genes of murine IL-12. The effects of paracrine secretion of IL-12 on tumor establishment and vaccination models were examined using the poorly immunogenic murine melanoma cell line (BL-6) in C57BL/6 mice. To determine the effects of IL-12 on tumor formation, nonirradiated BL-6 cells were inoculated s.c. into C57BL/6 mice admixed with NIH3T3 cells transfected with both subunits of mIL-12 (3T3-IL-12) or with cells transfected with only the neomycin phosphotransferase gene (3T3-Neo). Compared to mice given injections of BL-6 alone, the day of emergence of detectable tumors was significantly delayed in mice given injections of BL-6 admixed with 3T3-IL-12, but not in mice with BL-6 admixed with 3T3-Neo. Effectiveness in this system was related to the amount of IL-12 expressed by the 3T3-IL-12. To determine the ability of locally secreted IL-12 at the tumor site to induce antitumor immunity, 10(6) irradiated tumor cells mixed with 3T3-IL-12 or 3T3-Neo were injected as a vaccine, and the response to a tumor challenge was subsequently examined. With a tumor challenge of less than 1 x 10(5) nonirradiated BL-6 cells, significant delay of establishment of tumor was noted with a relatively small amount of IL-12 secretion (1.2 units/5 x 10(5) cells/48 h). Larger amounts of secreted IL-12 provided no additional therapeutic benefit. Histological examination of tumor inoculum with 3T3-IL-12 secreting a high level of IL-12 showed peritumoral accumulation of macrophages, a characteristic capsule around the tumor composed of palisades of fibroblasts, and decreased numbers of CD4+ cells in the tumor. These results suggest that local delivery of IL-12 inhibits tumor growth in a dose dependent manner but leads to the development of an antitumor immune response when IL-12 is expressed at the tumor site at the relatively small amount indicated above.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Fibroblastos/metabolismo , Interleucinas/metabolismo , Melanoma/prevención & control , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/metabolismo , Células 3T3 , Animales , Secuencia de Bases , División Celular/inmunología , Línea Celular , Inmunoterapia , Interleucina-12 , Interleucinas/genética , Kanamicina Quinasa , Melanoma/inmunología , Melanoma/patología , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA