Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 23 Suppl 8: viii22-7, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22918924

RESUMEN

The prognosis for men with metastatic, castration-resistant prostate cancer (CRPC) is limited, and patients have very few treatment options, particularly if the treatment failed with docetaxel (Taxotere). As a result, there is a requirement for novel approaches to therapy. Using immunotherapy to induce immune responses to prostate cancer in preclinical and clinical studies appears to be a valid therapeutic approach. In a pivotal phase III trial, treatment with sipuleucel-T, an autologous cellular vaccine consisting of activated antigen-presenting cells loaded with prostatic acid phosphatase (PAP), gave a median overall survival of 25.8 months compared with 21.7 months for placebo-treated patients, resulting in a 22% relative reduction in the risk of death. Based on these results, sipuleucel-T became the first therapeutic vaccine approved for any type of cancer in the USA. PROSTVAC(®)-VF, a poxvirus-based vaccine engineered to present prostate-specific antigen (PSA) and three immune costimulatory molecules, and GVAX, a vaccine consisting of two prostate cancer cell lines (LnCAP and PC3) and genetically modified to secrete granulocyte-macrophage colony-stimulating factor (GM-CSF), both showed promising results in phase II studies, although GVAX failed to meet its primary end point of overall survival when compared with docetaxel in a phase III study. T-cell modulation is another potential immunotherapeutic strategy for CRPC. Ipilimumab, an antibody against the cytotoxic T-lymphocyte-associated antigen-4, is being evaluated in phase I/II studies, both alone and in combination with chemotherapy, radiotherapy or GVAX, with activity in prostate cancer. CRPC is one of the few tumour types where immunotherapy is the current standard of care. Further research, however, will be necessary to improve antitumour responses and clinical benefits, including the use of novel combinatorial approaches.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Extractos de Tejidos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno CTLA-4/inmunología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Docetaxel , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Ipilimumab , Masculino , Tasa de Supervivencia , Taxoides/uso terapéutico , Resultado del Tratamiento
2.
Prostate Cancer Prostatic Dis ; 24(3): 871-879, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33746212

RESUMEN

BACKGROUND: In 2004 docetaxel was the first life-prolonging drug (LPD) registered for metastatic castration-resistant prostate cancer (mCRPC) patients. Between 2011 and 2014 new LPDs for mCRPC (cabazitaxel, abiraterone, enzalutamide, and radium-223) were introduced in the Netherlands. The objective of this study is to assess the impact of the introduction of new LPDs on treatment patterns and overall survival (OS) over time. PATIENTS AND METHODS: CRPC patients diagnosed in the years 2010-2016 in the observational, retrospective CAPRI registry (20 hospitals) were included and followed up to 2018. Two subgroups were analyzed: treatment-naïve patients (subgroup 1, n = 3600) and post-docetaxel patients (subgroup 2, n = 1355). RESULTS: In both subgroups, the use of any LPD increased: from 57% (2010-2011) to 69% (2014-2015) in subgroup 1 and from 65% (2011-2012) to 79% (2015-2016) in subgroup 2. Chemotherapy as first mCRPC-treatment (i.e., docetaxel) and first post-docetaxel treatment (i.e., cabazitaxel or docetaxel rechallenge) decreased (46-29% and 20-9% in subgroup 1 and 2, respectively), while the use of androgen-receptor targeting treatments (ART) increased from 11% to 39% and 46% to 64% in subgroup 1 and 2, respectively. In subgroup 1, median OS (mOS) from diagnosis CRPC increased from 28.5 months to 31.0 months (p = 0.196). In subgroup 2, mOS from progression on docetaxel increased from 7.9 months to 12.5 months (p < 0.001). After multiple imputations of missing values, in multivariable cox-regression analysis with known prognostic parameters, the treatment period was independent significant for OS in subgroup 1 (2014-2015 vs. 2010-2011 with HR 0.749, p < 0.001) and subgroup 2 (2015-2016 vs. 2011-2012 with HR 0.811, p = 0.037). CONCLUSION: Since 2010, a larger proportion of mCRPC patients was treated with LPDs, which was related to an increased mOS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Quimioradioterapia/mortalidad , Neoplasias de la Próstata Resistentes a la Castración/mortalidad , Radio (Elemento)/uso terapéutico , Anciano , Anciano de 80 o más Años , Androstenos/administración & dosificación , Benzamidas/administración & dosificación , Docetaxel/administración & dosificación , Estudios de Seguimiento , Humanos , Masculino , Nitrilos/administración & dosificación , Feniltiohidantoína/administración & dosificación , Pronóstico , Neoplasias de la Próstata Resistentes a la Castración/patología , Neoplasias de la Próstata Resistentes a la Castración/terapia , Estudios Retrospectivos , Tasa de Supervivencia , Taxoides/administración & dosificación
3.
Clin Transl Oncol ; 21(6): 774-780, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30465182

RESUMEN

BACKGROUND: Health-related quality of life (HRQoL) is an important issue in the rapidly evolving field of adjuvant treatment for stage III melanoma. Dendritic cell vaccination is one of the adjuvant forms of therapy currently investigated. METHODS: We enrolled adults with stage III melanoma to receive adjuvant dendritic cell vaccination after a complete radical lymph node dissection. HRQoL assessment was one of the secondary endpoints of this trial and investigated with the EORTC-QLQ-C30 questionnaire at baseline and week 26. RESULTS: Fifteen patients with a median age of 50 years were included in the study, with twelve evaluable patients on study at time of the second questionnaire. Global health status and role functioning improved clinically relevant with a mean difference of 15 (p = 0.010) and 26 points (p = 0.005), respectively. DISCUSSION: Despite the small number of patients, we found a clinically relevant improved global health status. Besides, compared to the other investigated therapies, toxicity of dendritic cell vaccination is low, which supports our finding. CONCLUSION: This is the first description of HRQoL in melanoma patients receiving dendritic cell vaccination. We show the expected improvement in global health status after surgical treatment of stage III melanoma. Thus, adjuvant dendritic cell vaccination does not seem to hamper this improvement, as shown in our small explorative study.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Células Dendríticas/trasplante , Inmunoterapia , Melanoma/terapia , Calidad de Vida , Adulto , Anciano , Células Dendríticas/inmunología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Melanoma/inmunología , Persona de Mediana Edad , Estadificación de Neoplasias , Estudios Prospectivos , Encuestas y Cuestionarios , Adulto Joven
4.
Crit Rev Oncol Hematol ; 122: 157-163, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29458784

RESUMEN

BACKGROUND: The purpose of this review was to assess the effectiveness of different strategies to implement physical activity during and after cancer treatment. DESIGN: We searched for studies containing strategies to implement physical activity in cancer care that meet the inclusion criteria of the Cochrane EPOC group. The primary outcome was physical activity uptake. We expressed the effectiveness of the strategies as the percentage of studies with improvement. RESULTS: Nine studies met the inclusion criteria. Patient groups doing physical activities via an implementation strategy had better outcomes than those receiving usual care: 83% of the studies showed improvement. Strategies showing significant improvement compared to usual care employed healthcare professionals to provide individual counselling or advice for exercise or interactive elements such as audit and feedback systems. When comparing the different strategies 1) interactive elements or 2) elements tailored to the needs of the patients had better physical activity uptake. CONCLUSIONS: Implementation strategies containing individual and interactive elements, tailored to the individual needs of patients, are more successful in improving physical activity uptake.


Asunto(s)
Ejercicio Físico/fisiología , Neoplasias/terapia , Humanos , Resultado del Tratamiento
5.
Oncol Res ; 16(10): 471-7, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18196871

RESUMEN

Multicellular tumor spheroids are used as a model to assess the efficacy of replicating oncolytic adenoviruses. As most assays used to assess cellular viability are unsuitable for oncolytic viruses because of ongoing viral replication, we have used positron emission tomography (PET) to sequentially determine the incorporation of 18F-labeled deoxyglucose (18F-DG) as a measure of viability and compared the results to more commonly used assays for measuring the effect of oncolytic therapy. Glioma monolayer cultures and spheroids were infected with wild-type replicating adenovirus and viability was measured by 18F-DG incorporation, WST-1 assay, crystal violet assay, and spheroid volume 2 to 10 days following infection. Results show that volume measurements in adenovirus-infected spheroids are confounded by the cytopathic effect occurring in infected cells. 18F-DG PET provides a useful method to assess small differences in cell number and viability following oncolytic viral therapy in glioma monolayer cultures and spheroids without the need for disintegration of these cultures. Moreover, using 18F-DG PET, repeated sequential measurements of spheroid viability can be made, decreasing the required number of spheroids per experiment. This is a valuable feature when using spheroids derived from limited amounts of patient material.


Asunto(s)
Adenoviridae/fisiología , Fluorodesoxiglucosa F18 , Glioma/diagnóstico por imagen , Glioma/terapia , Viroterapia Oncolítica/métodos , Glioma/genética , Glioma/virología , Humanos , Tomografía de Emisión de Positrones/métodos , Esferoides Celulares , Células Tumorales Cultivadas
6.
Leukemia ; 16(4): 650-7, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11960346

RESUMEN

The chemokine stromal cell-derived factor-1 (SDF-1) and its receptor CXCR-4 contribute to stem cell homing and may play a role in the trafficking of leukemic cells. Therefore, we analyzed migration across Transwell filters of cells derived from bone marrow (BM) or peripheral blood (PB) from 26 acute myeloid leukemia (AML) patients. The presence of the extracellular matrix protein fibronectin (FN) strongly enhanced the spontaneous and SDF-1-induced migration of leukemic PB and BM cells. No differences in spontaneous, SDF-1-induced migration or CXCR-4 expression were observed between the different AML subtypes. Subsequently, it was determined whether SDF-1 preferentially promoted migration of subsets of leukemic cells. Leukemic cells expressing CD34, CD38 and HLA-DR were preferentially migrating, whereas cells expressing CD14 and CD36 showed diminished migration. Analysis of paired PB and BM samples indicated that significantly higher SDF-1-induced migration was observed in AML for CD34(+) BM-derived cells compared to CD34(+) PB-derived cells, suggesting a role for SDF-1 in the anchoring of leukemic cells in the BM or other organs. The lower percentage of circulating leukemic blasts in patients with a relatively high level of SDF-1-induced migration also supports this hypothesis. In conclusion, we have shown that primary AML cells are migrating towards SDF-1 independent of the AML subtypes.


Asunto(s)
Movimiento Celular , Leucemia Mieloide/patología , Enfermedad Aguda , Adulto , Anciano , Anciano de 80 o más Años , Antígenos CD/metabolismo , Médula Ósea/patología , Ciclo Celular , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiocinas CXC/farmacología , Femenino , Fibronectinas/farmacología , Citometría de Flujo , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas , Humanos , Leucemia Mieloide/metabolismo , Masculino , Persona de Mediana Edad , Fenotipo , Receptores CXCR4/metabolismo , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología
7.
Leukemia ; 12(8): 1195-203, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9697873

RESUMEN

Hepatocyte growth factor (HGF), also known as scatter factor (SF), is produced by mesenchymal cells, including bone marrow (BM) stromal cells, and has mitogenic and motogenic effects on a variety of cell types. Recently, a role has been assigned to HGF/SF and its receptor, c-MET, in both normal and malignant hemopoiesis. We investigated the function of HGF/SF on hemopoietic mononuclear cells (MNC) from patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) with circulating blasts. In contrast to results with normal MNC, HGF/SF alone stimulated the proliferation and colony formation of MNC from these patients. MNC from some (4/13) of the AML patients also produced HGF/SF (0.1-0.2 ng/ml/day), while we could not detect HGF/SF in cultures from normal MNC. Furthermore, it appeared that HGF/SF induced migration of leukemic cells in Boyden using KG1a cells as a model for leukemic blasts. The membranes dividing the two compartments of the Boyden chambers were coated with fibronectin. HGF/SF significantly promoted migration in 3/5 samples of MDS patients and in 5/7 samples of AML patients. Supernatant of human BM stromal cells, which is chemoattractive for normal human hemopoietic progenitor cells, also promoted migration of MNC from 4/5 MDS patients and 6/7 AML patients. Since HGF/SF is one of the growth factors produced by BM stromal cells, a neutralizing antibody directed against HGF/SF was added to the BM stroma supernatant, which reduced migration significantly in 2/3 MDS and in 3/6 AML responders to BM stroma supernatant. In conclusion, HGF/SF promotes proliferation and migration of hemopoietic cells from AML and MDS patients in vitro and may therefore contribute to the malignant potential of these cells.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Leucemia Mieloide/metabolismo , Enfermedad Aguda , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Adhesión Celular , División Celular , Movimiento Celular , Ensayo de Unidades Formadoras de Colonias , Fibronectinas/metabolismo , Humanos , Técnicas In Vitro , Leucemia Mieloide/patología , Síndromes Mielodisplásicos/metabolismo , Síndromes Mielodisplásicos/patología , Células del Estroma/metabolismo
8.
Clin Cancer Res ; 7(3): 641-50, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11297260

RESUMEN

Adenoviral-mediated gene transfer is suboptimal in human glioma and limits in vivo gene therapy approaches. There is a need for targeted vectors able to enhance gene transfer into the tumor as well as to lower the viral load in the surrounding normal tissues. We evaluated primary human tumor samples by immunohistochemistry and fluorescence-activated cell sorter for expression of the Coxsackie-adenovirus receptor and other antigens with potential utility to redirect adenoviruses (Ads) to gliomas. In the majority of the samples, Coxsackie-adenovirus receptor expression was low. This correlated with inefficient gene transfer in vitro. Epidermal growth factor receptor (EGFR) and alpha(v)beta5 integrins were often highly, but heterogeneously, expressed. We hypothesized that these receptors, overexpressed in tumor but not in normal brain, could serve as independent binding sites for alternative pathways of infection with targeted Ads. We examined this, using Ads that expressed the luciferase reporter gene under the cytomegalovirus promoter. Targeting to the EGFR was performed with a single-chain bispecific antibody directed against the human EGFR and against the fiber knob of the Ad. Targeting to the alpha(v) integrins was performed by insertion of an integrin-binding sequence, RGD-4C, in the HI-loop of the Ad. Increased luciferase gene transfer in primary glioma cells was observed in 8 of 13 samples with EGFR-targeting (2-11 times enhancement; median, 6) and in all of the samples with RGD-targeting (2-42 times enhancement; median, 12). Combining the two targeting motifs further enhanced the gene transfer in primary glioma cells in an additive manner (3-56 times; median, 20). The double-targeted Ads also strongly augmented gene transfer into organotypic glioma spheroids. Conversely, gene transfer into normal brain explants was reduced dramatically using Ads targeted to the tumor. Our findings demonstrate the feasibility and benefit of binding multiple ligands to the adenoviral fiber knob. These vectors have a great potential for clinical use in the context of tumors that are usually heterogeneous for target antigen expression at the single-cell level.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , Técnicas de Transferencia de Gen , Glioma/metabolismo , Integrinas/metabolismo , Esferoides Celulares/metabolismo , Anticuerpos Biespecíficos/metabolismo , Antígenos CD/metabolismo , Encéfalo/embriología , Neoplasias Encefálicas/patología , Separación Celular , Enterovirus/genética , Citometría de Flujo , Glioblastoma/metabolismo , Glioblastoma/patología , Glioma/patología , Humanos , Inmunohistoquímica , Integrina alfaV , Luciferasas/genética , Luciferasas/metabolismo , Regiones Promotoras Genéticas , Células Tumorales Cultivadas
9.
Exp Hematol ; 27(12): 1806-14, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10641598

RESUMEN

Hematopoietic progenitor cells (CD34+ cells) migrate to the bone marrow after reinfusion into peripheral veins. Stromal cell-derived factor-1 (SDF-1) is a chemokine produced by bone marrow stromal cells that induces migration of CD34+ cells. In this study we compared spontaneous and SDF-1-induced migration of CD34+ cells from bone marrow (BM), peripheral blood (PB), and cord blood (CB) across Transwell filters. Under all circumstances, CB CD34+ cells showed significantly more migration than did BM or PB CD34+ cells. SDF-1 induced migration of BM CD34+ cells was higher than that of PB CD34+ cells, possibly due to differences in sensitivity towards SDF-1. Indeed, PB CD34+ cells showed a significantly lower expression of the receptor for SDF-1 (CXCR-4) than did BM and CB CD34+ cells. The sensitivity to SDF-1, as measured by migration towards different concentrations of SDF-1, was identical for BM and CB-derived CD34+ cells and correlated with their equal CXCR-4 receptor expression. Coating of the filters with the extracellular matrix protein fibronectin (FN) strongly enhanced the SDF-1-induced migration of PB CD34+ cells (2.5 times) and of BM CD34+ cells (1.5 times). SDF-1 induced migration of PB CD34+ cells over FN-coated filters was blocked by antibodies against beta1 integrins. Subsequently, analysis was performed to determine whether SDF-1 preferentially promoted migration of subsets of CD34+ cells. Actively cycling CD34+ cells, which were present in BM (14%) but hardly in PB (2.2%) or CB (1.2%), were found to migrate preferentially towards SDF-1. In the input, 14%+/-2.5% of the BM CD34+ cells were in G2/M and S phase, whereas in the migrated fraction 20%+/-5.7% of the cells were actively cycling (p < 0.05). We did not observe preferential migration of phenotypically recognizable primitive CD34+ subsets, despite the fact that CB CD34+ cells are thought to contain a higher percentage of immature subsets. In conclusion, the relatively lower migration of PB CD34+ cells seems to be due to a lower sensitivity towards SDF-1, and the higher migrational capacity of CB CD34+ cells, in comparison to BM and PB CD34+ cells, seems to have an as yet unknown intrinsic cause. The increased migration of CB CD34+ cells may favor homing of these cells to the bone marrow, which might reduce the number of cells required for hematological reconstitution after transplantation.


Asunto(s)
Médula Ósea , Movimiento Celular , Sangre Fetal/citología , Fibronectinas , Células Madre Hematopoyéticas/citología , Movilización de Célula Madre Hematopoyética , Humanos
10.
Exp Hematol ; 27(8): 1306-14, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10428507

RESUMEN

Homing of hematopoietic progenitor cells (HPC) to the bone marrow may be mediated by adhesion molecules specifically expressed on human bone marrow endothelial cells (HBMEC). This hypothesis suggests that HPC would preferentially bind to HBMEC compared to endothelial cells from other origins. In this study, HPC were allowed to adhere either to HBMEC cell lines or to human umbilical vein endothelial cells (HUVEC) in two different experimental set-ups. First, adherence was measured using a flow cytometric assay with three different colors identifying each cell population (HPC, HBMEC, HUVEC). HPC could adhere (in a competitive way) to the two endothelial cell lines under stirring conditions, which simulated adhesion under shear stress, as present in blood vessels. Because this assay requires relatively firm adhesion and the endothelial cells don't form a monolayer, we studied the same interactions under less stringent conditions. HPC were allowed to adhere to endothelial monolayers under gently rocking conditions. Differential adhesion of HPC to a set of endothelial cell lines did not correlate with the origin of the endothelial cells. Adhesion of HPC to both types of endothelial cells was inhibited in the presence of various combinations of monoclonal antibodies against the adhesion molecules VLA-4, CD18, and/or E-selectin. No indications were obtained for qualitative differences in the role of these molecules in adhesion of HPC to either HBMEC or HUVEC cell lines. In conclusion, no preferential adhesion of HPC to HBMEC compared to HUVEC cells was observed. This may be due to a lack of origin-specific differences between endothelial cells, implying that the specificity of homing is not regulated at the entrance of the bone marrow. Otherwise, the origin-specific differences between endothelial cells of different origins may be microenvironment-induced, rather then intrinsic, implying that care should be exercised with the use of endothelial cell lines in studies investigating the specificity of homing of HPC.


Asunto(s)
Células de la Médula Ósea/citología , Moléculas de Adhesión Celular/metabolismo , Quimiotaxis/fisiología , Endotelio Vascular/citología , Células Madre Hematopoyéticas/citología , Venas Umbilicales/citología , Anticuerpos Monoclonales/farmacología , Unión Competitiva , Antígenos CD18/inmunología , Antígenos CD18/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular/inmunología , Selectina E/inmunología , Selectina E/metabolismo , Endotelio/citología , Citometría de Flujo , Humanos , Integrina alfa4beta1 , Integrinas/antagonistas & inhibidores , Integrinas/inmunología , Integrinas/metabolismo , Especificidad de Órganos , Receptores Mensajeros de Linfocitos/antagonistas & inhibidores , Receptores Mensajeros de Linfocitos/inmunología , Receptores Mensajeros de Linfocitos/metabolismo
11.
Exp Hematol ; 26(9): 885-94, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9694510

RESUMEN

The fate of hematopoietic progenitor cells (HPCs) in the bone marrow (BM) microenvironment is determined by two different interactions: 1) they adhere (via integrins) to both extracellular matrix molecules and BM stromal cells; and 2) stromal cells produce cytokines that influence their survival, proliferation, differentiation, and mobilization. The ligands for the protein tyrosine kinase receptors c-KIT and FLT3/FLK2, stem cell factor (SCF), and FL are produced by BM stromal cells and are known to affect several facets of hematopoiesis. We studied another protein tyrosine kinase receptor, c-MET, and its ligand hepatocyte growth factor (HGF), also known as scatter factor (SF), which play a similar role in hematopoiesis. c-MET mRNA is expressed in immature human BM HPCs (CD34+CD33- or CD34+CD38-), but not in more mature HPCs (CD34+CD33+ or CD34+CD38+). The ligand HGF/SF is predominantly produced by BM stromal cells at both the mRNA and protein levels. We confirmed functionally that HGF/SF alone has no effect on proliferation of HPCs, but that when combined with granulocyte/macrophage colony-stimulating factor (GM-CSF) or interleukin-3 it acts as a synergistic proliferative factor, although not as potently as kit-ligand or FLT-3/FLK-2 ligand. Furthermore, HGF/SF promotes adhesion of HPCs to immobilized fibronectin. HGF/SF-induced adhesion to fibronectin is probably caused by activation of the integrins alpha4beta1 and alpha5beta1, insofar as we were able to block this interaction by using monoclonal blocking antibodies directed against these integrin subunits. Addition of the tyrosine-phosphorylation inhibitor genistein inhibited HGF/SF-induced adhesion, supporting the idea that HGF/SF-induced effects are the result of signaling via the receptor c-MET after ligand binding. The enhanced adhesion of HGF/SF to fibronectin proved to be beneficial for the maintenance of the colony-forming potential of HPCs. HGF/SF alone and especially in combination with fibronectin prolongs survival of GM colony-forming cells in liquid culture. Our data indicate that HGF/SF is a polyfunctional cytokine in the BM microenvironment. It is produced by human BM stromal cells and directly or indirectly promotes proliferation, adhesion, and survival of human HPCs.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/citología , Factor de Crecimiento de Hepatocito/fisiología , Adhesión Celular , División Celular , Ensayo de Unidades Formadoras de Colonias , ADN Complementario/genética , Sinergismo Farmacológico , Fibronectinas/metabolismo , Fibronectinas/farmacología , Expresión Génica , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Factor de Crecimiento de Hepatocito/biosíntesis , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/farmacología , Humanos , Integrina alfa4beta1 , Integrinas/fisiología , Interleucina-3/farmacología , Proteínas de la Membrana/farmacología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-met/biosíntesis , Proteínas Proto-Oncogénicas c-met/genética , Proteínas Proto-Oncogénicas c-met/fisiología , ARN Mensajero/biosíntesis , Receptores de Fibronectina/fisiología , Receptores Mensajeros de Linfocitos/fisiología , Factor de Células Madre/farmacología , Células del Estroma/metabolismo
12.
Cancer Gene Ther ; 7(8): 1197-9, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10975681

RESUMEN

The "First International Symposium on Genetic Anticancer Agents," which took place in Amsterdam on March 8-9, 2000, served as a forum to review the results of preclinical and clinical gene therapy studies for cancer endeavored to date. Despite the fact that gene therapy was initially conceptualized as an approach for inherited genetic disease, it is currently finding its widest employ for treating neoplastic disorders. In this regard, more than 70% of patients treated to date in human clinical gene therapy protocols have been in the context of anticancer regimens. Of note, the application of gene therapy for cancer has proceeded from the same rational basis as was originally conceptualized for inherited genetic disorders. Specifically, the molecular basis of these disorders is increasingly being understood, therapeutic genes are available, and alternative therapies are often lacking. Most recently, the field of gene therapy has enjoyed the realization of the first incontrovertible evidence of clinical benefit, for hemophilia and cardiovascular disease, in its first 15 years of human application. This recent recognition of the potential power of gene therapy, and the current lack of realizing such ends for neoplastic disease, has led to a reassessment of the field. Such a critical analysis is a necessary step in defining the means to progress the technology toward achieving the potential benefits of gene therapy for cancer.


Asunto(s)
Terapia Genética , Neoplasias/terapia , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia , Profármacos/uso terapéutico
13.
Cancer Gene Ther ; 7(6): 901-4, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10880021

RESUMEN

Recombinant adenoviral vectors are attractive in the context of cancer gene therapy because they are capable of delivering genes to a wide variety of tissues. The utility of adenoviruses is limited by their lack of specificity and by the absence of the receptor(s) for these viruses on many tumor cells. Redirecting adenoviral vectors to tissue- or tumor-specific targets can be achieved by using bispecific conjugates produced by chemical linkage of an anti-adenovirus antibody (Ab) and a ligand or Ab directed toward a specific target. To avoid the limitations of chemical conjugates, molecular conjugates of anti-fiber knob and ligand have been proposed. We present here a novel strategy that allows the production of recombinant bispecific single-chain Abs directed at cell surface molecules. A construct was made that encodes a neutralizing anti-adenovirus fiber single-chain Fv (scFv) Ab (S11) fused to a scFv Ab (425) directed against the epidermal growth factor receptor. The fusion protein markedly enhanced the infection efficiency of adenoviral vectors in epidermal growth factor receptor-expressing cell lines. The bispecific scFv could be purified and concentrated after binding of its 6His tag to a nickel column without significant loss of activity. This approach should permit the production of high quantities of active bispecific scFv for in vivo use. The universal design of the construct allows rapid screening for relevant specific scFv directed at cell surface antigens that can be incorporated into adenoviral targeting strategies.


Asunto(s)
Infecciones por Adenoviridae , Adenoviridae/genética , Anticuerpos Biespecíficos/genética , Anticuerpos Antivirales/genética , Marcación de Gen/métodos , Terapia Genética/métodos , Vectores Genéticos , Adenoviridae/inmunología , Animales , Western Blotting , Células COS/metabolismo , Células Cultivadas/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Fragmentos de Inmunoglobulinas/inmunología , Región Variable de Inmunoglobulina/inmunología , Proteínas Recombinantes de Fusión
14.
Cancer Gene Ther ; 8(5): 342-51, 2001 May.
Artículo en Inglés | MEDLINE | ID: mdl-11477454

RESUMEN

Application of recombinant adenoviral vectors for cancer gene therapy is currently limited due to lack of specificity for tumor cells. For gastric and esophageal adenocarcinoma, we present here that the relative abundant expression of the primary adenovirus receptor, coxsackie/adenovirus receptor (CAR), on normal epithelium compared to carcinoma favors the transduction of the epithelium. As such, to achieve specific transduction of cancer cells, targeting approaches are required that ablate the binding of the virus to CAR and redirect the virus to tumor-specific receptors. By immunohistochemistry and reverse transcriptase polymerase chain reaction assays, we demonstrate a marked difference in expression of the human epithelial cell adhesion molecule (EpCAM) between normal and (pre)malignant lesions of the stomach and esophagus. Based on this, we explored the feasibility of using EpCAM to achieve gastric and esophageal adenocarcinoma selective gene transfer. Adenoviral vectors redirected to EpCAM using bispecific antibodies against the adenovirus fiber-knob protein and EpCAM specifically infected gastric and esophageal cancer cell lines. Using primary human cells, an improved ratio of tumor transduction over normal epithelium transduction was accomplished by the EpCAM-targeted vectors. This study thus indicates that EpCAM-targeted adenoviral vectors may be useful for gastric and esophageal cancer-specific gene therapy in patients.


Asunto(s)
Adenocarcinoma/terapia , Adenoviridae/genética , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Neoplasias Esofágicas/terapia , Neoplasias Gástricas/terapia , Adenocarcinoma/metabolismo , Adenocarcinoma/virología , Cartilla de ADN/química , Molécula de Adhesión Celular Epitelial , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/virología , Marcación de Gen/métodos , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Técnicas para Inmunoenzimas , Adhesión en Parafina , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virología , Células Tumorales Cultivadas
15.
Transplantation ; 45(2): 301-7, 1988 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-3278419

RESUMEN

Complement-mediated lysis of (subsets of) T lymphocytes in bone marrow grafts is increasingly used to prevent acute graft-versus-host disease in human bone marrow transplant recipients, especially in case of major immunogenetic disparity between donor and recipient. Since T lymphocyte depletion has resulted in an increased frequency of allogeneic engraftment failures, its effect on hemopoietic reconstitution was measured in rhesus monkeys. The reactivity patterns of commonly used types of antihuman T lymphocyte monoclonal antibodies (MCAs) with rhesus monkey lymphocytes was analyzed using a double-label cytofluorometry technique and found to be very similar to those with human lymphocytes. The antibodies investigated included CAMPATH-1 (recognizing an antigen present on virtually all lymphocytes and monocytes), OKT4 + 4a (CD4, helper/inducer T lymphocytes), B9 (CD8, suppressor/cytotoxic T lymphocytes), WT-1 (CD7, pan-T), and anti-DR MCAs as stem cell toxic controls. Their possible toxicity to hemopoietic stem cells was studied by using a semiquantitative autologous regeneration assay. Cytotoxic lysis of cells in the bone marrow grafts reacting with the T lymphocyte purging MCAs did not result in delayed regeneration compared to untreated autologous grafts. It is concluded that T lymphocyte depletion using anti-T-lymphocyte MCAs does not influence the repopulating capacity of an autologous bone marrow graft.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Trasplante de Médula Ósea , Células Madre Hematopoyéticas/citología , Linfocitos T/inmunología , Animales , Médula Ósea/inmunología , Células de la Médula Ósea , División Celular , Ensayo de Unidades Formadoras de Colonias , Proteínas del Sistema Complemento/farmacología , Reacciones Cruzadas , Citotoxicidad Inmunológica , Células Madre Hematopoyéticas/inmunología , Humanos , Depleción Linfocítica/efectos adversos , Macaca mulatta , Linfocitos T/citología
16.
Expert Opin Biol Ther ; 2(8): 943-52, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12517272

RESUMEN

Standard therapies are not capable of curing patients with malignant glioma; more than 90% of patients die within 2 years after diagnosis. Gene therapy appeared as a promising new approach for this disease. However, results of clinical trials with replication deficient viral vectors were disappointing. The main reasons being poor transduction efficiency of adenovirus towards glioma cells and limited spread and distribution of the vector in the tumour. With the increasing knowledge of viral genetics and its functions, an attractive alternative tool to kill malignant glioma cells has been developed: Replicating adenovirus as an oncolytic agent. This type of therapy, also referred to as virotherapy, has the potential to overcome some of the limitations connected with replication deficient adenoviral vectors. In this review the authors describe the latest developments in strategies that are being used to create a tumour- or glioma-selective replicating adenovirus. Special attention is given to the methods of viral delivery to an infiltrating tumour in the brain, regarding optimal dose and toxicity. Furthermore, the role of conventional antitumour treatments, such as irradiation and chemotherapy, in enhancing the effect of virotherapy is being emphasised.


Asunto(s)
Adenoviridae , Neoplasias Encefálicas/terapia , Virus , Animales , Línea Celular Tumoral , Glioma/terapia , Humanos
17.
Bone Marrow Transplant ; 13(4): 441-7, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8019469

RESUMEN

Fluorescence in situ hybridization (FISH) enables the visualization of the X and Y chromosomes in nuclei of interphase cells using probes which recognize repetitive sequences on the X and Y chromosomes. The process of cell preparation, hybridization and visualization of the probes within the nuclei lasts between 16 and 24 h. Because of these features the method is ideal for monitoring engraftment after sex-mismatched bone marrow transplant (BMT). So far, the FISH technique has been applied to assess the degree of chimerism in either unseparated peripheral blood cells or unseparated bone marrow cells. However, for early detection of rejection or relapse, the degree of chimerism in subpopulations of cells is probably more informative. A new FISH protocol was developed with the aim of simultaneously visualizing the surface antigens by fluorescent antibodies and X or Y chromosomes with fluorescent probes. After development of the new FISH protocol, it was applied to patient samples after sex-mismatched BMT. The degree of chimerism was assessed comparing the results of the new FISH protocol with a 'standard' FISH protocol which is employed both in our and other laboratories to detect chromosomes in interphase nuclei. Both methods gave similar results. The new FISH protocol was applicable for the detection of Y and X chromosomes. The clinical relevance of this new FISH protocol is illustrated by the results obtained on peripheral blood samples of five BMT patients who were mixed chimeras after sex-mismatched BMT. A clear distinction between engraftment of T and B cells was noted. In some patients the T cells were of donor origin while the B cells were of host origin.


Asunto(s)
Antígenos de Superficie/análisis , Células Sanguíneas/química , Trasplante de Médula Ósea/patología , Técnica del Anticuerpo Fluorescente , Supervivencia de Injerto , Hibridación Fluorescente in Situ , Cromosoma X , Cromosoma Y , Adulto , Antígenos de Superficie/genética , Niño , Preescolar , Quimera , Femenino , Humanos , Síndromes de Inmunodeficiencia/terapia , Leucemia/terapia , Masculino , Talasemia beta/terapia
19.
Leuk Lymphoma ; 13(3-4): 215-21, 1994 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7914126

RESUMEN

Myelodysplastic syndromes (MDS) comprises a group of acquired hematological disorders which is characterized by a progressive peripheral blood pancytopenia of one or more cell lineages. A high percentage of blast cells in either bone marrow or peripheral blood predisposes for the transformation to acute myeloid leukemia. The clinical presentation with pancytopenia suggest that all cell lineages are affected by MDS. The first experiments with X-linked restriction fragment length polymorphism (RFLP) indicated that MDS is a stem cell disorder since the clonal deletions could be detected in all cell lineages. During the 1st decade several new molecular biological techniques such as polymerase chain reaction, and fluorescent in situ hybridization (FISH) were applied to study molecular aberrations in subpopulations of cells. Molecular aberrations in all subpopulations would indicate that MDS is a stem cell disorder. The clonal studies in MDS are equivocal. Studies involving the expression of chromosomal abnormalities (standard karyotyping and FISH) in different cell lineages suggest that the pluripotent stem cell is not affected in MDS since the lymphoid cells usually do not express the abnormal karyotype. Results obtained by RFLP vary widely. Some studies indicate that the lymphoid lineage is not involved, while other studies find a polyclonal expression of the polymorphic genes in lymphoid cells. One study using PCR demonstrated mutations in the ras-oncogenes in T-cells as well as myeloid cells, suggesting that a common ancestor of myeloid and lymphoid cells is affected by MDS. This review discusses the different experimental approaches carried out to solve the discussion whether MDS is a stem cell disorder.


Asunto(s)
Células Clonales/patología , Células Madre Hematopoyéticas/patología , Síndromes Mielodisplásicos/patología , Línea Celular Transformada , Aberraciones Cromosómicas , Herpesvirus Humano 4 , Humanos , Hibridación Fluorescente in Situ , Síndromes Mielodisplásicos/clasificación , Síndromes Mielodisplásicos/genética , Oncogenes , Reacción en Cadena de la Polimerasa , Polimorfismo de Longitud del Fragmento de Restricción
20.
J Control Release ; 87(1-3): 159-65, 2003 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-12618032

RESUMEN

Recombinant adenoviral vectors are promising reagents for therapeutic interventions in humans, including gene therapy for biologically complex diseases like cancer and cardiovascular diseases. In this regard, the major advantage of adenoviral vectors is their superior in vivo gene transfer efficiency on a wide spectrum of both dividing and non-dividing cell types. However, this broad tropism at the same time represents an important limitation for their use in therapeutic applications where specific gene transfer is required. This limitation may be overcome by using targeting approaches. In this regard, targeting may be achieved at three levels: transductional targeting, translational targeting and targeting of the expressed transgene. Here we describe our research efforts towards cancer specific gene therapy using these different targeting approaches. The results show that targeting of adenoviral vectors may be achieved using cancer specific cell surface molecules for transductional and transgene targeting or cancer specific promoters for transcriptional targeting. Combinations of these targeting approaches should result in optimized cancer specific gene therapy.


Asunto(s)
Adenoviridae/genética , Sistemas de Liberación de Medicamentos/métodos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Humanos , Neoplasias/genética , Docilidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA