Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
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
2.
Cancer Res ; 51(16): 4199-205, 1991 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-1868441

RESUMEN

A Phase I trial of tumor-infiltrating lymphocytes (TIL) expanded in vitro and administered on Days 1 and 8, with or without continuous infusion recombinant interleukin 2 (rIL-2) in 25 patients with metastatic renal cell carcinoma, was conducted. Eighteen of the 25 eligible patients were treated with TIL and escalating doses of rIL-2 (0.0, 3.0, 4.5 x 10(6) units/m2) on Days 1 to 5 and 8 to 12. Dose-limiting toxicity was pulmonary, and the maximum tolerated dose of rIL-2 was 3.0 x 10(6) units/m2. No clinical responses were observed. Immunological monitoring of peripheral blood lymphocytes demonstrated significant increases in CD3+ and CD56+ cells, including the activated T-cell subsets. Phenotypic analysis of cultured TILs demonstrated significant heterogeneity and the presence of CD3+CD4+ and CD3+CD8+ T-cells, with CD3-CD56+ and CD3+CD56+ populations also present. The majority of cultured TILs expressed HLA-DR and CD45RO, with a variable number expressing CD25. The rIL-2-expanded TILs possessed cytotoxicity against allogeneic and autologous tumor, with cytolytic activity against only autologous tumor seen in one patient. Results demonstrate that in vitro expansion of TILs is possible, but further studies are needed to define the biology of TILs in renal cancer and to isolate and expand tumor-specific T-cells.


Asunto(s)
Carcinoma de Células Renales/terapia , Inmunoterapia Adoptiva/efectos adversos , Interleucina-2/toxicidad , Neoplasias Renales/terapia , Linfocitos Infiltrantes de Tumor/inmunología , Adulto , Antígenos CD/análisis , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/patología , Línea Celular , Células Cultivadas , Citotoxicidad Inmunológica , Evaluación de Medicamentos , Antígenos HLA-DR/inmunología , Humanos , Interleucina-2/uso terapéutico , Neoplasias Renales/inmunología , Neoplasias Renales/patología , Metástasis de la Neoplasia , Estadificación de Neoplasias , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/toxicidad , Subgrupos de Linfocitos T/inmunología
3.
Cancer Res ; 54(20): 5424-9, 1994 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-7923175

RESUMEN

Recent data suggest that the poor induction of a T-cell response to human renal cell carcinoma (RCC) may be related to alterations in signal transduction pathways. We report that T cells from RCC patients have two alterations in kappa B motif-specific DNA-binding activity. The first alteration involves the constitutive expression of substantial kappa B-binding activity in nuclear extracts, which was observed in the electrophoretic mobility shift assay. The magnitude of kappa B activity in unstimulated patient T cells was similar to that observed in T cells from normal individuals that had been activated in vitro. On the basis of Western blotting experiments using antibodies to kappa B/Rel family proteins, the kappa B-binding activity constitutively expressed in T cells from RCC patients is composed mostly of the NF-kappa B1 (p50) subunit. The second abnormality in kappa B-binding activity in T cells from these patients is that RelA, a member of the Rel homology family which is part of the normal NF-kappa B complex, was not induced in the nucleus following activation. Western blotting analysis did not detect any RelA in nuclear extracts either before or after stimulation of T cells. The altered kappa B-binding activity in T cells from RCC patients may impair their capacity to respond normally to various stimuli.


Asunto(s)
Carcinoma de Células Renales/inmunología , ADN de Neoplasias/metabolismo , Neoplasias Renales/inmunología , FN-kappa B/metabolismo , Linfocitos T/inmunología , Secuencia de Bases , Western Blotting , Humanos , Datos de Secuencia Molecular , Transducción de Señal , Linfocitos T/metabolismo
4.
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
5.
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
6.
Clin Cancer Res ; 5(10): 2780-9, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10537342

RESUMEN

Interleukin 12 (IL-12) is known to play an important role in the development of an antitumor response. Its activity has been shown to be dependent upon the intermediate production of IFN-gamma and the influx into the tumor of CD8 lymphocytes. In a murine model, tumor regression induced by IL-12 treatment correlated with IFN-gamma, IP-10, and Mig expression in the tumor bed and was abrogated by antibodies to both chemokines. Here we examined the effects of rHuIL-12 on IFN-gamma and CXC chemokine gene expression in patients with renal cell carcinoma (RCC) in an attempt to determine whether a similar series of molecular events leading to IL-12-mediated tumor regression in mice is also detectable in humans. As in the murine RENCA model, cultured RCC cells themselves could be induced by IFN-gamma to synthesize IP-10 and Mig mRNA. Explanted RCC produced IFN-gamma and IP-10 mRNA in response to IL-12 treatment, which was consistent with the finding that biopsied RCC tumors from IL-12-treated patients also variably expressed augmented levels of those molecules after therapy. Although Mig mRNA was present in the majority of biopsied tumors prior to treatment, both the Mig and IP-10 chemokines as well as IFN-gamma were induced in the peripheral blood mononuclear cells of IL-12-treated patients. Skin biopsies of IL-12-treated patients also all synthesized IP-10 mRNA. This study demonstrates that recombinant human IL-12 therapy of patients with RCC has the potential to induce the expression of gene products within the tumor bed that may contribute to the development of a successful antitumor response.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Quimiocinas CXC/biosíntesis , Péptidos y Proteínas de Señalización Intercelular , Interferón gamma/biosíntesis , Interleucina-12/farmacología , Neoplasias Renales/tratamiento farmacológico , Animales , Quimiocina CXCL10 , Quimiocina CXCL9 , Quimiocinas CXC/genética , Humanos , Ratones , ARN Mensajero/análisis , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas
7.
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
8.
Clin Cancer Res ; 2(2): 347-57, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9816178

RESUMEN

We conducted a Phase I trial of s.c. recombinant human interleukin 3 (rhIL-3) to evaluate the toxicity, maximal tolerated dose, pharmacokinetics, and in vivo biological effects of this cytokine. Thirty-one patients with refractory cancer were entered into the study between November 1991 and June 1993. Therapy consisted of s.c. rhIL-3 daily for 15 days administered to cohorts of three to nine patients at dose levels of 60-4000 microgram/m2/day. Cycles were repeated at intervals of 28 days. Seventy-five cycles of rhIL-3 were administered (median, two per patient) and the maximal tolerated dose was 2000 microgram/m2/day. Toxicity was moderate, with most patients developing chills, fever, and myalgia. Dose-limiting toxicity consisted of diarrhea (two patients) and headache (one patient). Hematological effects of rhIL-3 included significant dose-related increases of WBC (P < 0.001), neutrophils (P < 0.001), and eosinophils (P < 0.001). Platelet counts and absolute lymphocyte numbers also increased. Various CD3(+) lymphocyte subsets increased; however, lytic activity (natural killer and lymphokine-activated killer) of peripheral blood lymphocytes was not enhanced. Serum levels of the soluble IL-2 receptor increased in a dose-related fashion, and IL-2-induced lymphocyte proliferation also was increased variably. Pharmacokinetic studies were performed in 13 patients, and area under the curve and maximal concentration values increased with increasing rhIL-3 dose levels (P < 0.001) and correlated with maximal changes from baseline in WBC, neutrophils, and eosinophils. rhIL-3 antibodies were detected in 8% of patients by day 29 of cycle 1 but were not neutralizing. rhIL-3 is well tolerated when administered s.c. and has reproducible hematological and immunological effects. The pleiotropic effects of this cytokine on various in vivo biological parameters were demonstrated clearly. Further studies of its immunoregulatory effects are warranted.


Asunto(s)
Interleucina-3/efectos adversos , Neoplasias/terapia , Adulto , Anciano , Recuento de Células Sanguíneas/efectos de los fármacos , Femenino , Humanos , Inyecciones Subcutáneas , Interleucina-3/administración & dosificación , Interleucina-3/farmacocinética , Interleucina-6/sangre , Activación de Linfocitos/efectos de los fármacos , Masculino , Persona de Mediana Edad , Neoplasias/sangre , Neoplasias/inmunología , Receptores de Interleucina-2/análisis , Proteínas Recombinantes/efectos adversos
9.
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
10.
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
11.
J Cancer Res Clin Oncol ; 127(10): 619-24, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11599798

RESUMEN

PURPOSE: Some limitations of effective therapy in multiple myeloma include the low growth fraction of the malignant plasma cells, multi-drug resistance, and the presence of other concurrent diseases in this patient population. A phase I study was conducted to evaluate the toxicity of granulocyte macrophage colony stimulating factor (GM-CSF) in myeloma patients as well as the potential effect on the plasma cell labeling index (PCLI). Relapsed patients with multiple myeloma were eligible. METHODS: The first phase of this trial assessed the toxicity (including the effect on disease progression) of escalating doses (125-500 microg/m2 SC, days 1-5) of GM-CSF, and the effects of this cytokine on PCLI. Patients whose PCLI doubled and increased to > or = 1.7% were treated with chemotherapy including cyclophosphamide, vincristine, prednisone, and GM-CSF. Twenty-two patients were enrolled. RESULTS: The toxicity of GM-CSF was mild, and no dose-limiting side effects were seen. Twenty-five percent of patients (5/20) achieved the target PCLI, and 4/5 proceeded to receive chemotherapy. No relationship of GM-CSF dose to increases of the PCLI was noted. All patients who received chemotherapy responded. CONCLUSIONS: GM-CSF has acceptable toxicity in patients with multiple myeloma and produced increases of PCLI in selected individuals. Further studies of GM-CSF alone or in combination with chemotherapy are indicated.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Inyecciones Intravenosas/efectos adversos , Mieloma Múltiple/tratamiento farmacológico , Alanina Transaminasa/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Aspartato Aminotransferasas/sangre , Femenino , Humanos , Masculino , Mieloma Múltiple/patología , Estadificación de Neoplasias , Selección de Paciente , Proteínas Recombinantes
12.
Oncogene ; 32(33): 3829-39, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22964639

RESUMEN

We report on the role of hexamethylene-bis-acetamide-inducible protein 1 (HEXIM1) as an inhibitor of metastasis. HEXIM1 expression is decreased in human metastatic breast cancers when compared with matched primary breast tumors. Similarly we observed decreased expression of HEXIM1 in lung metastasis when compared with primary mammary tumors in a mouse model of metastatic breast cancer, the polyoma middle T antigen (PyMT) transgenic mouse. Re-expression of HEXIM1 (through transgene expression or localized delivery of a small molecule inducer of HEXIM1 expression, hexamethylene-bis-acetamide) in PyMT mice resulted in inhibition of metastasis to the lung. Our present studies indicate that HEXIM1 downregulation of HIF(-)1α protein allows not only for inhibition of vascular endothelial growth factor-regulated angiogenesis, but also for inhibition of compensatory pro-angiogenic pathways and recruitment of bone marrow-derived cells (BMDCs). Another novel finding is that HEXIM1 inhibits cell migration and invasion that can be partly attributed to decreased membrane localization of the 67 kDa laminin receptor, 67LR, and inhibition of the functional interaction of 67LR with laminin. Thus, HEXIM1 re-expression in breast cancer has therapeutic advantages by simultaneously targeting more than one pathway involved in angiogenesis and metastasis. Our results also support the potential for HEXIM1 to indirectly act on multiple cell types to suppress metastatic cancer.


Asunto(s)
Neoplasias Mamarias Experimentales/metabolismo , Neovascularización Patológica/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Femenino , Citometría de Flujo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Inmunoprecipitación , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Células MCF-7 , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Invasividad Neoplásica/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas de Unión al ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Espectrometría de Masas en Tándem , Análisis de Matrices Tisulares , Técnicas del Sistema de Dos Híbridos
15.
J Immunol ; 163(2): 590-8, 1999 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-10395645

RESUMEN

NF-kappa B is involved in the transcriptional control of various genes that act as extrinsic and intrinsic survival factors for T cells. Our findings show that suppression of NF-kappa B activity with cell-permeable SN50 peptide, which masks the nuclear localization sequence of NF-kappa B1 dimers and prevents their nuclear localization, induces apoptosis in resting normal human PBL. Inhibition of NF-kappa B resulted in the externalization of phosphatidylserine, induction of DNA breaks, and morphological changes consistent with apoptosis. DNA fragmentation was efficiently blocked by the caspase inhibitor Z-VAD-fmk and partially blocked by Ac-DEVD-fmk, suggesting that SN50-mediated apoptosis is caspase-dependent. Interestingly, apoptosis induced by NF-kappa B suppression, in contrast to that induced by TPEN (N,N,N',N'-tetrakis [2-pyridylmethyl]ethylenediamine) or soluble Fas ligand (CD95), was observed in the absence of active death effector proteases caspase-1-like (IL-1 converting enzyme), caspase-3-like (CPP32/Yama/apopain), and caspase-6-like and without cleavage of caspase-3 substrates poly(ADP-ribose) polymerase and DNA fragmentation factor-45. These findings suggest either low level of activation is required or that different caspases are involved. Preactivation of T cells resulting in NF-kappa B nuclear translocation protected cells from SN50-induced apoptosis. Our findings demonstrate an essential role of NF-kappa B in survival of naive PBL.


Asunto(s)
Apoptosis/inmunología , Caspasa 1/metabolismo , Caspasas/metabolismo , Caspasas/fisiología , FN-kappa B/antagonistas & inhibidores , Linfocitos T/enzimología , Linfocitos T/metabolismo , Apoptosis/efectos de los fármacos , Caspasa 3 , Caspasa 6 , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Activación Enzimática/inmunología , Humanos , Inmunosupresores/farmacología , Interfase/inmunología , Activación de Linfocitos/efectos de los fármacos , FN-kappa B/metabolismo , FN-kappa B/fisiología , Péptidos/fisiología , Fosfatidilserinas/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
16.
Int J Cancer ; 53(6): 941-7, 1993 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8473051

RESUMEN

Previous studies have documented the effects of IL2 on the growth and effector function of tumor-infiltrating lymphocytes (TIL) in cancer patients. Since IL7 is known to induce T- and NK-cell responses in the peripheral blood, we examined the immuno-enhancing effects of IL7 on TIL derived from human renal-cell carcinoma (RCC). Whereas IL2 induced the growth of freshly isolated TIL in vitro, IL7 was ineffective alone and failed to increase the total number of cells proliferating to IL2. However, IL7 did provide a proliferative signal to TIL that were initially expanded in culture with either IL2 or IL2/IL7 for 2 weeks. IL7 also induced the proliferation of CD4+ and CD8+ TIL lines that have specificity for RCC. The proliferative response induced by IL7 was independent of IL2, since anti-IL2 antibodies did not block IL7-induced proliferation of TIL. IL7 did cooperate with anti-CD3 stimulation for the induction of proliferation; however, the magnitude of this interaction was variable and the response usually additive. In addition, IL7 synergized with anti-CD3 to induce the secretion of IFN gamma from short-term-cultured TIL and from a TIL line. Although IL7 did not promote the development of a tumor-specific T-cell response from IL2-expanded TIL, IL7 enhanced lymphokine-activated killer (LAK) activity from some short-term-cultured TIL. These results illustrate that IL7 can potentiate the growth and production of IFN gamma from RCC-reactive TIL and, to a lesser extent, enhance IL2-induced LAK activity of TIL.


Asunto(s)
Carcinoma de Células Renales/inmunología , Interleucina-7/farmacología , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/fisiología , Linfocitos T Reguladores/efectos de los fármacos , Carcinoma de Células Renales/tratamiento farmacológico , División Celular/efectos de los fármacos , Humanos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucina-2/farmacología , Neoplasias Renales/tratamiento farmacológico , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/inmunología , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Reguladores/inmunología
17.
J Immunother ; 22(1): 71-9, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9924702

RESUMEN

Interleukin-12 (IL-12) is a heterodimeric cytokine that enhances the cytolytic activity, proliferation, and interferon-gamma (IFN-gamma) production by T lymphocytes and natural killer cells, and has significant antitumor activity in a variety of murine tumor models. The induction of interferon (IFN)-gamma by IL-12 in tumor-bearing mice plays an important role in its antitumor activity. We therefore examined the effects of IL-12 on IFN-gamma production by T cells derived from patients with renal cell carcinoma (RCC), including freshly isolated tumor infiltrating lymphocytes (T-TIL), matched peripheral blood T cells (T-PBL), and RCC-specific TIL lines. IL-12 alone induced IFN-gamma secretion by T cells from normal individuals and appeared to act synergistically with either IL-2 or anti-CD3 antibody. In contrast, it failed to stimulate significant IFN-gamma secretion by T-PBL and T-TIL from RCC patients. This unresponsive state in T-PBL appeared selective because IFN-gamma was produced when cells were stimulated with either phytohemagglutinin or anti-CD3 antibody. Moreover, costimulation through the T-cell receptor (TCR)/CD3 complex or with IL-2 made T-PBL from RCC patients responsive to IL-12, possibly secondary to the upregulation of IL-12R (beta chain). A selective loss of IL-12-dependent production of IFN-gamma was also consistently observed in two of three established RCC-specific TIL lines. Although these cell lines did not respond to any concentration of IL-12, they did produce IFN-gamma after ligation of the TCR/CD3 or stimulation with IL-2, IL-12 also acted either syngeristically or additively with IL-2, anti-CD3 antibody, or autologous tumor cells to induce IFN-gamma production. The observed decreases in IFN-gamma production in response to IL-12 may have a negative effect on the development of T-cell immunity. The clinical importance of these findings during in vivo administration of IL-12 remains to be determined.


Asunto(s)
Carcinoma de Células Renales/inmunología , Interferón gamma/biosíntesis , Interleucina-12/farmacología , Neoplasias Renales/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T/inmunología , Carcinoma de Células Renales/tratamiento farmacológico , Separación Celular , ADN/biosíntesis , Citometría de Flujo , Humanos , Interleucina-2/farmacología , Neoplasias Renales/tratamiento farmacológico , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Muromonab-CD3/inmunología , Fitohemaglutininas/farmacología , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12 , Proteínas Recombinantes/farmacología , Linfocitos T/efectos de los fármacos , Células Tumorales Cultivadas
18.
Int J Cancer ; 45(1): 119-24, 1990 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-2298495

RESUMEN

TIL were cultured from human renal-cell carcinoma (RCC) in 1,000 U/ml rIL2 and restimulated with autologous tumor in efforts to establish which conditions would best expand the number of lymphocytes cytotoxic for autologous tumor. Greater cell yields resulted from multiple restimulations of TIL with autologous tumor. In most instances, these TIL lysed autologous tumor better than TIL grown in rIL2 alone. Enhanced proliferation was seen also after restimulation of TIL with allogeneic RCC as well as with tumor cells of non-renal origin. Although in some instances lysis of autologous tumor appeared to be specific, restimulation with autologous tumor did not consistently result in the generation of specific cytolytic T cells. Attempts to culture more specific cytolytic T cells by using 50 U/ml rIL2 were successful in expanding TIL with enhanced lytic activity; however, this activity was not specific for autologous tumor. The phenotype of the tumor-restimulated TIL generally did not change. In most of the TIL cultures, CD3+CD4+ cells were predominant with low numbers of CD3+Leu19+ cells and minimal numbers of CD3-Leu19+ cells. Thus, the cytotoxic response to tumor was mediated by T cells and not NK cells. Overall, our data indicate that restimulation of TIL with autologous tumor may be beneficial for growing larger numbers of cells which have increased lytic activity and for prolonging the presence of lytic activity among the expanded TIL.


Asunto(s)
Carcinoma de Células Renales/inmunología , Neoplasias Renales/inmunología , Activación de Linfocitos/inmunología , Linfocitos/inmunología , Adulto , Anciano , Carcinoma de Células Renales/patología , División Celular/inmunología , Línea Celular/inmunología , Línea Celular/patología , Separación Celular/métodos , Citotoxicidad Inmunológica/inmunología , Femenino , Humanos , Neoplasias Renales/patología , Linfocitos/citología , Masculino , Persona de Mediana Edad , Fenotipo , Células Tumorales Cultivadas/inmunología , Células Tumorales Cultivadas/patología
19.
J Immunother Emphasis Tumor Immunol ; 15(2): 91-104, 1994 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-7907918

RESUMEN

The detection of T cells with specificity for human renal cell carcinoma (RCC) has been difficult to document. In an attempt to improve our identification of RCC-reactive T cells, tumor-infiltrating lymphocytes (TIL) were expanded in interleukin-2/interleukin-4 (IL-2/IL-4) and then separated into CD4+ and CD8+ subsets using antibody-coated biomagnetic beads. TIL grown in IL-2/IL-4 expanded to greater numbers than TIL grown in IL-2 alone. From 16 patients in whom subset separation was performed, three CD4+ and three CD8+ TIL consistently had specificity for RCC that was detected by cytotoxicity, proliferation, or interferon-gamma (IFN-gamma) production. Four of the six lines were derived from the IL-2/IL-4 cultures. Two CD8+ TIL lines displayed specific lytic activity, lysing the autologous tumor but not allogeneic RCC or nonrenal tumors. Moreover, the lytic activity of these lines was blocked by anti-CD3 antibody, suggesting that tumor recognition was through the TCR/CD3 complex. Two additional TIL lines showed preferential lysis of RCC because they were cytotoxic for autologous tumor and one or more allogeneic RCC but not other tumor types. Two nonlytic CD4+ lines as well as the two CD8+ lines that were specifically lytic also produced IFN-gamma in response to the autologous tumor but not allogeneic RCC. Although these TIL lines produce IFN-gamma when stimulated with tumor alone, the addition of 5 U/ml of IL-2 significantly enhanced IFN-gamma secretion. The four TIL lines that showed specificity for RCC in terms of IFN-gamma production also had enhanced proliferation to the autologous RCC plus IL-2 but not to multiple allogeneic RCC plus IL-2. These studies demonstrate that TIL from RCC patients contain both CD4+ and CD8+ T cells that have specificity for RCC. In addition to cytotoxicity, specificity to RCC can be defined by IFN-gamma production and proliferation.


Asunto(s)
Carcinoma de Células Renales/inmunología , Citotoxicidad Inmunológica/fisiología , Interferón gamma/metabolismo , Activación de Linfocitos/fisiología , Linfocitos Infiltrantes de Tumor/fisiología , Linfocitos T CD4-Positivos , Antígenos CD8 , Carcinoma de Células Renales/secundario , Humanos , Técnicas In Vitro , Interleucina-2/fisiología , Interleucina-4/fisiología , Neoplasias Renales/inmunología , Subgrupos de Linfocitos T
20.
Cancer Immunol Immunother ; 41(3): 175-84, 1995 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-7553687

RESUMEN

T cells infiltrating (T-TIL) B cell non-Hodgkin's lymphomas (NHL) are thought to represent a local host response to the tumor. However, tumor progression in the presence of this T cell infiltrate suggests that the T-TIL may be functionally impaired. To address this issue we determined whether response to stimulation of T-TIL from 25 patients with NHL through the T cell receptor (TCR/CD3) and the interleukin-2 (IL-2) receptor (IL-2R) was intact, since activation of these receptors is important for proliferation and cytokine production. Our results demonstrate defects in response to stimulation via TCR/CD3 and the IL-2R in T-TIL cells from patients with NHL that were not observed with T cells from the peripheral blood. T-TIL showed minimal proliferation to anti-CD3 and only modest proliferation to IL-2 alone or when combined with anti-CD3. Moreover, cytokine production in T-TIL was impaired since stimulation through the TCR/CD3 complex did not induce mRNA for interferon gamma (IFN gamma), IL-2, IL-4 or IL-10. The functional unresponsiveness of these cells may be linked to altered signalling through the TCR/CD3 since an abnormal tyrosine phosphorylation pattern was detected in T-TIL after stimulation with anti-CD3.


Asunto(s)
Activación de Linfocitos/fisiología , Linfocitos Infiltrantes de Tumor/inmunología , Linfoma de Células B/inmunología , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Interleucina-2/fisiología , Adulto , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Secuencia de Bases , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-2/farmacología , Ionomicina/farmacología , Ionóforos/farmacología , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/fisiología , Linfoma de Células B/metabolismo , Datos de Secuencia Molecular , Fosforilación , Tirosina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA