Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Pharm ; 12(1): 34-45, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25412253

RESUMEN

UNLABELLED: In vitro studies of ocular bioavailability of active pharmaceutical ingredients (API) from colloidal drug delivery systems do not consider physiological shear stress generated by eyelid wiping and tear flow. The present study introduces a live cell imaging approach which enables the investigation of model drug uptake from various formulations under shear stress by using custom-made microchannels for the cultivation of human corneal epithelial cells (HCE-T). Coumarin-6 (C-6) was used as a model API incorporated into solid lipid nanoparticles and liposomes, and as an aqueous crystalline suspension. Confocal laser scanning microscopy visualized C-6 uptake into HCE-T cells in a time-resolved manner with an applied shear stress of 0.1 Pa. Static conditions were also studied for comparative purposes. Additionally, solid lipid nanoparticles (SLN) were labeled with a fluorescent phospholipid to check whether C-6 uptake was associated with SLN incorporation into the cells. RESULTS: Intact SLN were not incorporated into the cells, i.e., C-6 was passively redistributed from SLN to lipophilic cellular compartments. C-6 was enriched up to a given limit in HCE-T cells within 5 min of contact with the dispersions both under static and under flow conditions. The C-6 delivery rate from liposomes was superior to that from SLN whereby the suspension exhibited the lowest rate. C-6 release rates were comparable for static and flow conditions. Alternate flushing with formulations and buffer revealed that cells accumulated C-6. The results suggest that combining microfluidics with live cell imaging provides a valuable option for in vitro studies of ocular drug delivery.


Asunto(s)
Córnea/efectos de los fármacos , Cumarinas/química , Células Epiteliales/efectos de los fármacos , Nanopartículas/química , Tiazoles/química , Disponibilidad Biológica , Línea Celular , Línea Celular Tumoral , Supervivencia Celular , Córnea/metabolismo , Cristalización , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Células Epiteliales/citología , Ojo/efectos de los fármacos , Fluorometría/métodos , Humanos , Lípidos/química , Liposomas/química , Liposomas/metabolismo , Micelas , Técnicas Analíticas Microfluídicas , Microfluídica , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Fluorescente
2.
Inhal Toxicol ; 25(9): 536-43, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23905970

RESUMEN

The aim of this study was to investigate the potential cytotoxicity of solid lipid nanoparticles (SLN) loaded with sildenafil. The SLNs were tested as a new drug delivery system (DDS) for the inhalable treatment of pulmonary hypertension in human lungs. Solubility of sildenafil in SLN lipid matrix (30:70 phospholipid:triglyceride) was determined to 1% sildenafil base and 0.1% sildenafil citrate, respectively. Sildenafil-loaded SLN with particle size of approximately 180 nm and monomodal particle size distribution were successfully manufactured using a novel microchannel homogenization method and were stable up to three months. Sildenafil-loaded SLN were then used in in vitro and ex vivo models representing lung and heart tissue. For in vitro models, human alveolar epithelial cell line (A459) and mouse heart endothelium cell line (MHEC5-T) were used. For ex vivo models, rat precision cut lung slices (PCLS) and rat heart slices (PCHS) were used. All the models were treated with plain SLN and sildenafil-loaded SLN in a concentration range of 0-5000 µg/ml of lipid matrix. The toxicity was evaluated in vitro and ex vivo by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Median lethal dose 50% (LD50) values for A549 cells and PCLS were found to be in the range of 1200-1900 µg/ml while for MHEC5-T cells and precision cut heart slices values were found between 1500 and 2800 µg/ml. PCHS showed slightly higher LD50 values in comparison to PCLS. Considering the toxicological aspects, sildenafil-loaded SLN could have potential in the treatment of pulmonary hypertension via inhalation route.


Asunto(s)
Portadores de Fármacos/toxicidad , Nanopartículas/toxicidad , Inhibidores de Fosfodiesterasa 5/toxicidad , Piperazinas/toxicidad , Sulfonas/toxicidad , Animales , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Femenino , Humanos , Técnicas In Vitro , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Miocardio/patología , Nanopartículas/química , Fosfatidilcolinas/química , Inhibidores de Fosfodiesterasa 5/química , Piperazinas/química , Purinas/química , Purinas/toxicidad , Ratas , Ratas Wistar , Citrato de Sildenafil , Solubilidad , Sulfonas/química , Triglicéridos/química
3.
Science ; 218(4571): 475-7, 1982 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-6981846

RESUMEN

Mouse and human placental tissue contains a large number of mononuclear phagocytes. These cells, isolated from placenta, were shown to produce the multifaceted immune factor interleukin-1. Activity in the supernatants of 48-hour mononuclear phagocyte cultures was associated with a 12,000- to 18,000-dalton protein, consistent with known interleukin-1 characteristics. Stimulation of phagocytosis with latex beads increased the production and release of interleukin-1 from these placental cells, which may be a useful source of this protein.


Asunto(s)
Interleucina-1/biosíntesis , Fagocitos/inmunología , Placenta/inmunología , Animales , Células Cultivadas , Medios de Cultivo , Humanos , Interleucina-2/análisis , Ratones , Placenta/citología
4.
Eur J Pharm Biopharm ; 131: 224-231, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30149060

RESUMEN

Antisolvent precipitation of poorly water-soluble drugs is a promising formulation technique to synthesize amorphous nanoparticles. The dissolution behavior of these nanoparticles is improved because of the high specific surface area and the amorphous state, leading to an enhanced bioavailability of the drug molecules. Nevertheless, stabilization of precipitated drug nanoparticles against agglomeration and recrystallization, which constitutes a key issue for further processing steps, has turned out to be a major challenge. For that reason, the present study presents a synthesis method to produce long-term stable amorphous ibuprofen nanoparticles via antisolvent precipitation. To reach this goal, a new precipitation method was developed: antisolvent melt precipitation (AMP). Formulation strategies (e.g. varying fraction of stabilizer) as well as process parameters (e.g. temperature) were under study to estimate their influence on particle size, size distribution, crystallinity, morphology and stability of synthesized drug nanoparticles.


Asunto(s)
Antiinflamatorios no Esteroideos/química , Ibuprofeno/química , Algoritmos , Cristalización , Composición de Medicamentos , Estabilidad de Medicamentos , Nanopartículas , Tamaño de la Partícula , Solubilidad , Solventes , Temperatura
5.
J Clin Invest ; 104(6): 769-76, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10491412

RESUMEN

Activation of the transcription factor nuclear factor-kappaB (NFkappaB) is impaired in T cells from patients with renal cell carcinomas (RCCs). In circulating T cells from a subset of patients with RCCs, the suppression of NFkappaB binding activity is downstream from the stimulus-induced degradation of the cytoplasmic factor IkappaBalpha. Tumor-derived soluble products from cultured RCC explants inhibit NFkappaB activity in T cells from healthy volunteers, despite a normal level of stimulus-induced IkappaBalpha degradation in these cells. The inhibitory agent has several features characteristic of a ganglioside, including sensitivity to neuraminidase but not protease treatment; hydrophobicity; and molecular weight less than 3 kDa. Indeed, we detected gangliosides in supernatants from RCC explants and not from adjacent normal kidney tissue. Gangliosides prepared from RCC supernatants, as well as the purified bovine gangliosides G(m1) and G(d1a), suppressed NFkappaB binding activity in T cells and reduced expression of the cytokines IL-2 and IFN-gamma. Taken together, our findings suggest that tumor-derived gangliosides may blunt antitumor immune responses in patients with RCCs.


Asunto(s)
Carcinoma de Células Renales/inmunología , Gangliósidos/farmacología , Proteínas I-kappa B , Inmunosupresores/farmacología , Neoplasias Renales/inmunología , FN-kappa B/antagonistas & inhibidores , Linfocitos T/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Linfocitos T/metabolismo
6.
J Natl Cancer Inst ; 82(2): 124-8, 1990 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-2152945

RESUMEN

To better understand the modulatory effects of interleukin-2 (IL-2) on lymphocyte proliferation, we examined the clonality of the in vitro T-cell response by Southern blot hybridization. Tumor-infiltrating lymphocytes (TILs) grown in the presence of IL-2 for 15-26 days had detectable T-cell receptor beta-chain gene rearrangements, which indicated oligoclonal enhancement in culture in four of nine TIL samples. In contrast, none of 11 uncultured TIL samples had detectable gene rearrangements. Lack of detection in at least three of the five negative, cultured TIL samples could be explained by increased numbers of natural killer cells. We hypothesize that the oligoclonal expansion noted results from the enhanced response of immune-primed T cells to IL-2.


Asunto(s)
Carcinoma de Células Renales/inmunología , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T/efectos de los fármacos , Interleucina-2/farmacología , Neoplasias Renales/inmunología , Linfocitos T/efectos de los fármacos , Southern Blotting , Separación Celular , Células Cultivadas , Células Clonales , ADN de Neoplasias/análisis , Citometría de Flujo , Humanos , Recuento de Leucocitos , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología
7.
Cancer Res ; 50(4): 1176-82, 1990 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-2297766

RESUMEN

Studies from our laboratory and others have demonstrated that alpha-interferon (IFN alpha) can regulate natural killer cells and lymphokine-activated killer (LAK) cell activation. In vitro experiments have shown that IFN alpha has differential effects on both natural killer cells and LAK activity when combined with interleukin 2 (IL2); IFN alpha synergized with IL2 to augment natural killer cells activity while it suppressed the IL2-induced LAK response. Here we demonstrated that IFN alpha A/D can also regulate IL2-induced LAK activity in vivo with enhanced or suppressed activity depending on the IFN alpha A/D dose. The enhanced response was observed with the combination when 80,000 units/day of IFN alpha A/D were used and was detectable in the spleen, lung, and peritoneum. When a high dose of IFN alpha A/D was combined with IL2, a moderate reduction in LAK activity was noted in the spleen and peritoneum. In contrast, a high dose IFN alpha A/D augmented IL2-induced LAK activity in the lung even though it reduced the level of cellular infiltration. We have also evaluated the effect that IL2, IFN alpha A/D, and IL2 plus IFN alpha A/D have on the frequency of LAK precursors in the spleen and lung using limiting dilution analysis. Treatment of normal mice with IL2 alone increased the frequency of LAK precursor (LAKp) in the lung. This increase was associated with an infiltration of Thy-1+, asialo-GM1+, Lyt-2- lymphocytes into the lungs. Moreover, treatment with IL2 plus IFN alpha A/D enhanced the frequency of LAKp over that observed with IL2 alone. Treatment with the combination did not change the phenotype of LAKp in the lung from that seen with IL2. The increase in LAKp frequency induced by the combined treatment may not be a direct effect of IFN alpha A/D on precursor cells since IFN alpha A/D alone did not increase the frequency of LAKp in vivo or in vitro when added to limiting dilution analysis cultures. In contrast to what occurred in the lung, a consistent increase in LAKp was not seen in the spleen after treatment with IL2 or with the combination, although LAK activity was observed. These results demonstrated that in addition to inducing lytic activity from LAK effectors in vivo, IL2 treatment increased the number of precursor cells within the lung. Moreover, IFN alpha A/D in combination with IL2 influenced the level of LAKp in situ.


Asunto(s)
Interferón Tipo I/farmacología , Interleucina-2/farmacología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Pulmón/citología , Bazo/citología , Animales , Células Cultivadas , Interacciones Farmacológicas , Humanos , Hígado/citología , Hígado/inmunología , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes , Bazo/inmunología , Factores de Tiempo
8.
Cancer Res ; 53(23): 5613-6, 1993 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8242613

RESUMEN

Cancer patients and mice bearing tumors develop a progressive immunosuppression manifested by a decreased delayed-type hypersensitivity, decreased T-cell lytic activity, diminished production of lymphokines, and a reduced T-cell proliferative response. The mechanisms underlying these changes are incompletely understood. We recently reported the presence of marked alterations in signal transduction in T-cells from mice bearing long-term (28-day) tumours. We hypothesized that a soluble product produced by the tumor or resulting from the immune response to tumor might be responsible for inducing the changes in T-cells. Tumor-infiltrating lymphocytes from patients with renal cell carcinoma tested here showed, in 10 of 11 cases, a marked decrease in the expression of the T-cell receptor zeta chain and in p56lck tyrosine kinase. The presence of major alterations in the tumor-infiltrating lymphocytes with only minor changes in the peripheral blood leukocyte T-cells supports the notion that the defects are induced by exposure to tumor. These results suggest that tumor-infiltrating lymphocytes may be compromised in their antitumor efficacy in patients with renal cell cancer.


Asunto(s)
Carcinoma de Células Renales/química , Neoplasias Renales/química , Linfocitos Infiltrantes de Tumor/química , Proteínas de la Membrana/análisis , Proteínas Proto-Oncogénicas/análisis , Receptores de Antígenos de Linfocitos T/análisis , Carcinoma de Células Renales/inmunología , Humanos , Neoplasias Renales/inmunología , Activación de Linfocitos , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito , Linfocitos Infiltrantes de Tumor/inmunología
9.
Cancer Res ; 59(12): 2838-42, 1999 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-10383143

RESUMEN

The antitumor effect of immuno- and chemotherapeutic agents is executed through stimulation of apoptotic programs in susceptible cells. Apoptosis induced in tumor cells requires activation of members of the caspase family of proteases. Deficient expression or activation of caspases may account in part for the failure of many current anticancer therapies. However, recent studies suggest that cell death can proceed in the absence of caspases. We investigated the susceptibility of human renal cell carcinoma (RCC) lines to two distinct modes of cell death, apoptosis and necrosis. RCC lines displayed almost complete resistance to apoptosis in response to the intracellular zinc chelator, N,N,N'N'-tetrakis (2-pyridylmethyl) ethylenediamine (TPEN), which instead induced dramatic accumulation of nonapoptotic necrotic cells. Conversely, TPEN was a potent inducer of apoptosis in caspase-competent normal kidney cells (NK-72) and Jurkat T lymphocytes. Resistance to apoptosis in RCC lines correlated with almost complete loss of caspase-3 expression and variable down-regulation of caspase-7, caspase-8, and caspase-10. These data may explain the resistance of RCC to drugs inducing apoptosis and have important consequences for further attempts to manipulate tumor cell death.


Asunto(s)
Carcinoma de Células Renales/patología , Caspasas/metabolismo , Neoplasias Renales/patología , Apoptosis , Carcinoma de Células Renales/enzimología , Caspasas/deficiencia , Inhibidores de la Colinesterasa/farmacología , Etilenodiaminas/farmacología , Humanos , Células Jurkat , Neoplasias Renales/enzimología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/patología , Necrosis , Linfocitos T/efectos de los fármacos , Linfocitos T/patología , Células Tumorales Cultivadas
10.
Cancer Res ; 50(8): 2363-70, 1990 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-2107973

RESUMEN

Previously we showed that IL2 expanded tumor-infiltrating lymphocytes (TILs) from renal cell carcinoma mediated non-major histocompatibility complex-restricted cytotoxicity. Phenotypic analysis showed that cultured TILs were composed mostly of T-lymphocytes with varying numbers of CD4+, CD8+, and CD56+ (Leu19+) populations. Here we compared the cytolytic activity of the two predominant TIL subsets, CD3+CD4+ and CD3+CD8+, to that of the CD56+ populations. Using magnetic beads coated with antibodies to either CD4 or CD8, CD3+CD4+, and CD3+CD8+ TILs were isolated in a highly enriched form (greater than 92%) and could be expanded for over 40 days in vitro with 1000 units/ml IL2. In a 4-h 51Cr release assay the CD4+ and CD8+ TILs showed minimal lytic activity, whereas unseparated cells exhibited significant levels of non-major histocompatibility complex-restricted cytotoxicity. The lytic activity seen in the 4-h assay with unseparated TILs appeared to be related to the presence of CD56+ populations. With one exception none of the purified CD4+ or CD8+ TILs expressed any significant levels of CD56, while the unseparated TILs contained varying numbers of CD3+CD56+ and CD3-CD56+ populations. Cell-sorting experiments verified that the CD56+ populations were responsible for most of the lytic activity in 4 h even though CD3+CD56- cells represented the predominant cell type. Although CD3+CD56- TILs were minimally lytic in 4 h, we show here that both CD3+CD4+ and CD3+CD8+ subsets displayed substantial cytotoxicity in long-term assays. In the 18-h 51Cr release assay 5 of 6 CD4+ and 2 of 3 CD8+ TILs were lytic for the autologous tumor. In two cases, restimulation with the autologous tumor induced augmented cytolytic activity of TIL subsets and in one case induced lytic activity in 4 h. The cytotoxic activity of TIL subsets was further examined using a 72-h assay in which TILs were cocultured with a confluent layer of tumor cells. The degree of cytotoxicity was quantitated by measuring the amount of crystal violet dye that was incorporated by tumor cells which remained after the incubation period. CD4+ and CD8+ TILs typically caused greater than a 50% reduction of tumor cells in 3 days and the level of reduction was increased when IL2 was added to the cultures. All the CD4+ and CD8+ subset preparations were cytotoxic in the 3-day assay even though some were not lytic for certain targets in the 18-h 51Cr release assay.(ABSTRACT TRUNCATED AT 400 WORDS)


Asunto(s)
Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Antígenos CD4/análisis , Carcinoma de Células Renales/inmunología , Citotoxicidad Inmunológica , Neoplasias Renales/inmunología , Linfocitos T/inmunología , Adulto , Anciano , Antígenos CD8 , Células Cultivadas , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Humanos , Interleucina-2/inmunología , Interleucina-2/farmacología , Células Asesinas Activadas por Linfocinas/inmunología , Masculino , Persona de Mediana Edad , Fenotipo , Linfocitos T/efectos de los fármacos
11.
Crit Rev Immunol ; 21(1-3): 215-48, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11642606

RESUMEN

The T cells of many cancer patients are naturally sensitized to tumor-associated antigens (Ag), or they can readily be sensitized with vaccine maneuvers. In melanoma patients, the adoptive transfer of such T cells can often be causally linked to the objective regression of established tumors. So far, few patients have shown sustained clinical benefit from such therapy, but preclinical mouse studies have now clearly delineated the hurdles that must be overcome to render T-cell-based antitumor therapy effective. Contrary to earlier expectations, it is now established that remarkably potent CD4+ and CD8+ pre-effector T cells are naturally sensitized even in mice bearing progressive, weakly immunogenic tumors. However, such T cells often display signal transduction impairments as a consequence of the tumor environment, which limit their acquisition of optimal effector function. Extracorporealization and culture of these tumor-sensitized T cells with appropriate activation stimuli not only restores normal signal transduction, but also confers resolute effector activity that can often sustain tumor rejection upon reinfusion. In mouse studies, the L-selectin(low) fraction of T cells in tumor-draining lymph nodes (TDLN) constitutes the potent pre-effector population and comprises both CD4+ and helper-independent CD8+ T cells. Appropriate in vitro activation confers an apparently unrestricted trafficking capacity to this fraction, and even the ability to proliferate within the tumor bed, leading to unprecedented tumor rejection at anatomic sites (e.g., subcutaneous and intracranial) that were historically refractory to such treatment. Such results underscore the surprising capacity of appropriately activated effector T cells to withstand the immunosuppressive, tolerogenic, and apoptotic influences of the typical tumor environment. Given the increasingly appreciated and critical communications between T cells and host Ag-presenting cells (APC), which cross-present tumor Ag, it is likely that dendritic cell-based vaccine maneuvers that promote sensitization of T1-committed L-selectin(low) antitumor T cells will play an increasingly important role in adoptive therapy strategies.


Asunto(s)
Inmunoterapia Adoptiva , Neoplasias/terapia , Linfocitos T/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/inmunología , Comunicación Celular , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Antígenos de Histocompatibilidad Clase I/análisis , Humanos , Tolerancia Inmunológica , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Modelos Animales , Neoplasias/inmunología
12.
Clin Cancer Res ; 7(10): 3276-81, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11595725

RESUMEN

The antitumor effect of T cells is executed either through CD95 or Perforin (PFN)/Granzyme B (GrB) pathways. Induction of apoptosis by either mode requires activation of caspase family members. However, recent studies have suggested that cell death can proceed in the absence of caspase induction and apoptotic events. We investigated the contribution of CD95 and PFN/GrB-mediated cytotoxicity to apoptotic and necrotic mechanisms of cell death in human renal cell carcinoma. Although freshly isolated and cultured tumors expressed CD95 on their surface, they were resistant to CD95-mediated apoptosis. CD95 resistance coincided with decreased levels of FADD protein and diminished caspase-3-like activity. In contrast, we demonstrated that tumor cell death mediated by PFN/GrB can be achieved in the absence of functional caspase activity and is accompanied by a dramatic accumulation of nonapoptotic necrotic cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Apoptosis/efectos de los fármacos , Carcinoma de Células Renales/patología , Linfocitos T Citotóxicos/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/metabolismo , Proteínas Portadoras/metabolismo , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Resistencia a Antineoplásicos , Activación Enzimática/efectos de los fármacos , Proteína Ligando Fas , Proteína de Dominio de Muerte Asociada a Fas , Granzimas , Humanos , Células Jurkat , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacología , Necrosis , Perforina , Proteínas Citotóxicas Formadoras de Poros , Serina Endopeptidasas/farmacología , Células Tumorales Cultivadas , Receptor fas/inmunología , Receptor fas/metabolismo
13.
Clin Cancer Res ; 7(3 Suppl): 940s-946s, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11300495

RESUMEN

Antitumor immunity fails to adequately develop in many cancer patients, including those with renal cell carcinoma (RCC). A number of different mechanisms have been proposed to explain the immune dysfunction observed in cancer patient T cells. Here we show that T cells from RCC patients display increased sensitivity to apoptosis. Tumor-infiltrating lymphocytes (TILs) display the most profound sensitivity, because 10-15% of those cells are apoptotic when assessed by terminal deoxynucleotidyltransferase-mediated nick end labeling in situ, and the number of apoptotic TILs further increases after 24 h of culture. Peripheral blood T cells from RCC patients are not directly apoptotic, although T lymphocytes derived from 40% of those individuals undergo activation-induced cell death (AICD) upon in vitro stimulation with phorbol myristate acetate and ionomycin. This is in contrast to T cells from normal individuals, which are resistant to AICD. TILs and peripheral blood T cells from RCC patients also exhibit impaired activation of the transcription factor, nuclear factor (NF)-kappaB. Additional findings presented here indicate that the heightened sensitivity of patient T cells to apoptosis may be tumor induced, because supernatants from RCC explants sensitize, and in some instances directly induce, normal T cells to apoptosis. These same supernatants also inhibit NF-kappaB activation. RCC-derived gangliosides may represent one soluble tumor product capable of sensitizing T cells to apoptosis. Pretreatment with neuraminidase, but not proteinase K, abrogated the suppressive effects of tumor supernatants on both NF-kappaB activation and apoptosis. Additionally, gangliosides isolated from tumor supernatants not only inhibited NF-kappaB activation but also sensitized T cells to AICD. These findings demonstrate that tumor-derived soluble products, including gangliosides, may contribute to the immune dysfunction of T cells by altering their sensitivity to apoptosis.


Asunto(s)
Apoptosis , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Neoplasias Renales/inmunología , Neoplasias Renales/patología , FN-kappa B/fisiología , Núcleo Celular/metabolismo , Fragmentación del ADN , Activación Enzimática , Gangliósidos/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Ionomicina/farmacología , Ionóforos/farmacología , Acetato de Tetradecanoilforbol , Factores de Tiempo
14.
Clin Cancer Res ; 4(10): 2337-47, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9796963

RESUMEN

Studies have demonstrated abnormalities of the CD3/T-cell antigen receptor (TCR) and pathways of signal transduction in T lymphocytes from animals and patients with advanced malignancy. Diminished expression of TCRzeta and p56(lck) that are associated with the TCR and reduced nuclear localization of RelA containing nuclear factor kappaB (NFkappaB) complexes have been noted. These defects have been described in T cells from patients with malignant melanoma, renal cell carcinoma (RCC), ovarian cancer, and colorectal cancer. Preliminary observations also indicate possible correlation with clinical variables such as stage in selected instances. To further characterize altered expression of TCRzeta, p56(lck), and impaired activation of NFkappaB, T lymphocytes were obtained from 65 patients with RCC, the majority of whom were receiving combination cytokine therapy [interleukin (IL)-2, IFN alpha-containing regimens] and 37 control individuals. In 29 of these patients, levels of TCRzeta and p56(lck) were determined by Western blots of T-cell lysates and semiquantitated using densitometry. Relative levels were then correlated with a series of clinical variables including response to therapy, performance status, survival, disease sites, age, and others. In another group of 28 patients (three individuals from the first group), the frequency of abnormal NFkappaB activation was studied using electrophoretic mobility shift assays after activation of T cells with phorbol myristate acetate/ionomycin or anti-CD3 monoclonal antibody. Changes in these signaling molecules during cytokine treatment were also investigated. TCRzeta and p56(lck) were detected in the peripheral blood T cells in 27 of 29 patients, and overall, reduced levels were noted visually in 12 of 29 (41%) and 13 of 29 (45%) individuals, respectively. When levels were semiquantitated using densitometry, significant decreases of TCRzeta (P = 0.029) and p56(lck) (P = 0.029) but not CD3epsilon (P = 0.131), compared with control levels, were found. In patients treated with IL-2/IFN alpha-based therapy, relative levels of TCRzeta increased significantly (P = 0.002) on day 15 of cycle one compared with the baseline. Correlations of TCRzeta or p56(lck) levels with response or disease variables, except for lower TCRzeta levels (P < 0.001) in the presence of bone metastases, were not found. Abnormal NFkappaB activation after stimulation with phorbol myristate acetate/ionomycin and/or anti-CD3 monoclonal antibody was found in 59% of patients (17 of 28) and was not accounted for by the advanced age of the study cohort. Activation of NFkappaB in peripheral blood T cells was inducible during cytokine therapy in four of six individuals who displayed impaired NFkappaB activity prior to therapy. Moreover, impaired activation of NFkappaB does not appear linked to a reduction of TCRzeta expression, because in five patients, normal TCRzeta levels were present although kappaB binding was not inducible. In the majority of patients with advanced RCC, peripheral blood T cells express TCRzeta and p56(lck), and in a subset, reduced levels of these TCRzeta associated molecules are seen that may increase during cytokine-based therapy. Abnormal activation of NFkappaB is also present in >50% of patients and may also revert to normal during IL-2/IFN alpha-based treatment. This alteration in NFkappaB activation occurred in the presence of normal expression of TCRzeta-associated signaling elements. The clinical significance of these findings remains unclear.


Asunto(s)
Carcinoma de Células Renales/inmunología , Citocinas/uso terapéutico , Neoplasias Renales/inmunología , Transducción de Señal , Linfocitos T/metabolismo , Carcinoma de Células Renales/terapia , Humanos , Neoplasias Renales/terapia , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/análisis , Proteínas de la Membrana/análisis , FN-kappa B/metabolismo , Receptores de Antígenos de Linfocitos T/análisis
15.
Clin Cancer Res ; 5(5): 1219-29, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10353760

RESUMEN

Tumors may escape immune recognition and destruction through the induction of apoptosis in activated T lymphocytes. Results from several laboratories suggest that FasL (L/CD95L) expression in tumors may be responsible for this process. In this study of patients with renal cell carcinoma (RCC), we provide evidence for two mechanisms of T-cell apoptosis. One mechanism involves the induction of apoptosis via FasL expression in tumor cells. This is supported by several observations, including the fact that tumor cells in situ as well as cultured cell lines expressed FasL mRNA and protein by a variety of techniques. The FasL in RCC is functional because in coculture experiments, FasL+ tumors induced apoptosis in Fas-sensitive Jurkat T cells and in activated peripheral blood T cells but not in resting peripheral blood T cells. Most importantly, antibody to FasL partially blocked apoptosis of the activated T cells. Moreover, Fas was expressed by T cells derived from the peripheral blood (53% median) and tumor (44.3% median) of RCC patients. Finally, in situ staining for DNA breaks demonstrated apoptosis in a subset of T cells infiltrating renal tumors. These studies also identified a second mechanism of apoptosis in RCC patient peripheral T cells. Whereas these cells did not display DNA breaks when freshly isolated or after culture for 24 h in medium, peripheral blood T cells from RCC patients underwent activation-induced cell death after stimulation with either phorbol 12-myristate 13-acetate/ionomycin or anti-CD3/CD28 antibodies. Apoptosis mediated by exposure to FasL in tumor cells or through T-cell activation may contribute to the failure of RCC patients to develop an effective T-cell-mediated antitumor response.


Asunto(s)
Apoptosis/fisiología , Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/citología , Glicoproteínas de Membrana/fisiología , Proteínas de Neoplasias/fisiología , Linfocitos T Citotóxicos/citología , Apoptosis/efectos de los fármacos , Células Sanguíneas/inmunología , Carcinoma de Células Renales/sangre , Fragmentación del ADN , Proteína Ligando Fas , Humanos , Etiquetado Corte-Fin in Situ , Ionomicina/farmacología , Células Jurkat/inmunología , Neoplasias Renales/sangre , Activación de Linfocitos , Linfocitos Infiltrantes de Tumor/inmunología , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , Muromonab-CD3/farmacología , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , Linfocitos T Citotóxicos/inmunología , Acetato de Tetradecanoilforbol/farmacología , Células Tumorales Cultivadas , Receptor fas/fisiología
16.
J Immunother (1991) ; 11(1): 1-11, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1734943

RESUMEN

Human renal cell carcinoma (RCC) is one of the tumors most sensitive to immunotherapy and in that regard it is similar to malignant melanoma. Clinical studies reporting responses to therapy suggested that a host immune response may be involved in the antitumor activity induced by immunotherapy in these tumors. Although detection of a specific T cell response to melanoma has been well documented, this has not been the case for RCC. The lytic response of interleukin-2 (IL-2) cultured tumor-infiltrating lymphocytes (TILs) from RCC has been nonspecific. However, in this report we describe a CD8+ TIL line derived from a primary RCC tumor that displays specificity for the autologous tumor. This line is lytic for autologous RCC but does not lyse autologous lymphoblasts, allogeneic RCC, or tumor cell lines of other histologic types. It also proliferates specifically to the autologous tumor in the absence of exogenous IL-2. However, the addition of low dose IL-2 to the cultures can significantly augment its proliferative response. When stimulated with autologous RCC but not allogeneic RCC the CD8+ line will produce interferon-gamma (IFN-gamma). It appears that recognition of RCC by this TIL line is through the TCR/CD3 complex because anti-CD3 antibody blocks the lytic activity, proliferation and IFN-gamma production of the line in response to the autologous tumor. Additional studies illustrate that cytotoxic T lymphocytes with apparent specificity for the autologous tumor are present in unseparated cultured TILs that can be detected by clonal analysis. Collectively these results suggest that there is a specific T cell response to human RCC.


Asunto(s)
Antígenos CD8/análisis , Interferón gamma/análisis , Linfocitos Infiltrantes de Tumor/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Citotóxicos/inmunología , Carcinoma , Línea Celular , Separación Celular , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Neoplasias Renales , Células Tumorales Cultivadas
17.
Am J Trop Med Hyg ; 28(4): 756-62, 1979 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-464196

RESUMEN

Zymosan depletion of serum complement in guinea pigs rendered them highly resistant to lesion by Loxosceles reclusa spider venom. Guinea pigs deficient in C4 of the complement system are as sensitive to the venom as normal guinea pigs. The injection of 35 micrograms of whole recluse venom intradermally into guinea pigs lowered their complement level by 35.7%. Brown recluse spider venom in concentrations as slight as 0.02 micrograms protein/ml can totally inactivate one CH50 of guinea pig complement in vitro. Bee, scorpion, and other spider venoms had no influence on the hemolytic titer of complement. Fractionation of recluse spider venom by Sephadex G-200 filtration separated the complement-inactivating property of the venom into three major regions which could be distinguished on the basis of heat stability as well as size. None was neutralized by antivenom. Polyacrylamide gel electrophoresis of venom resolved the complement inactivators into five fractions. Complement inactivated by whole venom or the Sephadex fractions could be restored to hemolytic activity by supplements of fresh serum but not by heat-inactivated serum, pure C3, pure C5, or C3 and C5 in combination.


Asunto(s)
Venenos de Artrópodos/farmacología , Proteínas Inactivadoras de Complemento , Venenos de Araña/farmacología , Animales , Antivenenos/farmacología , Relación Dosis-Respuesta a Droga , Cobayas , Hemólisis , Calor , Venenos de Araña/análisis
18.
J Virol Methods ; 1(3): 167-77, 1980.
Artículo en Inglés | MEDLINE | ID: mdl-6168645

RESUMEN

Interferon (IF) treatment of murine cells chronically producing infectious leukemia virus results in a decreased yield of extracellular virus. We have utilized this principle to develop a useful semi-microassay for IF. The assay incorporates the desirable features of both virus yield-reduction and plaque-reduction assays, yet has few of the undesirable features associated with many IF assays. Being a semi-micro assay, it is amenable to the screening of multiple samples for IF and is economical with respect to cell culture materials, reagents and amount of sample required for testing.


Asunto(s)
Bioensayo/métodos , Interferones/análisis , Ensayo de Placa Viral , Animales , Línea Celular , Efecto Citopatogénico Viral , Interferones/farmacología , Ratones , Virus de la Leucemia Murina de Moloney/crecimiento & desarrollo
19.
Urology ; 33(2): 106-9, 1989 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2783793

RESUMEN

Interleukin-2-stimulated lymphocytes or lymphokine-activated killer cells (LAK) have been shown to have anti-tumor activity which is not present in the fresh untreated peripheral blood lymphocytes. This activity has been evaluated in our laboratory using renal cell carcinoma as a target tumor. Two of 8 patients demonstrated significant lysis of autologous tumor while an additional 3 patients lysed the target cells but to a less significant degree. It is not clear why the LAK phenomenon against autologous tumors occurs in vitro in some patients and not others. By identifying the response of patients in vitro, one might be able to select an appropriate population for meaningful clinical trials.


Asunto(s)
Carcinoma de Células Renales/inmunología , Inmunización Pasiva , Interleucina-2/inmunología , Neoplasias Renales/inmunología , Células Asesinas Naturales/inmunología , Linfocitos/inmunología , Línea Celular , Femenino , Humanos , Técnicas In Vitro , Masculino , Células Tumorales Cultivadas
20.
Anticancer Res ; 12(3): 921-5, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1622150

RESUMEN

Development of multidrug-resistance (MDR) remains a major cause of failure in the treatment of cancer with chemotherapeutic agents. In our efforts to explore alternative treatment regimens for multidrug-resistant tumors we have examined the sensitivity of MDR tumor cell lines to lymphokine activated killer (LAK) cells. Adriamycin (ADM) resistant B16-BL6 melanoma, L1210 and P388 leukemic cell lines were tested for sensitivity to lysis by LAK cells in vitro. While ADM-resistant B16-BL6 and L1210 sublines were found to exhibit at least 2-fold greater susceptibility to lysis by LAK cells, sensitivity of ADM-resistant P388 cell was similar to that of parental cells. Since ADM-resistant B16-BL6 cells were efficiently lysed by LAK cells in vitro, the efficacy of therapy with LAK cells against the ADM-resistant B16-BL6 subline in vivo was evaluated. Compared to mice bearing parental B16-BL6 tumor cells, the adoptive transfer of LAK cells and rIL2 significantly reduced formation of experimental metastases (P less than 0.009) and extended median survival time (P less than 0.001) of mice bearing ADM-resistant B16-BL6 tumor cells. Results suggest that immunotherapy with LAK cells and rIL2 may be a useful modality in the treatment of cancers with the MDR phenotype.


Asunto(s)
Doxorrubicina/farmacología , Inmunoterapia Adoptiva , Interleucina-2/uso terapéutico , Células Asesinas Activadas por Linfocinas/inmunología , Neoplasias Pulmonares/secundario , Melanoma Experimental/terapia , Animales , Células Clonales , Citotoxicidad Inmunológica , Resistencia a Medicamentos , Células Asesinas Activadas por Linfocinas/trasplante , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA