Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Invest New Drugs ; 19(4): 311-5, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11561690

RESUMEN

A phase II trial of gemcitabine (Gemzar), a nucleoside analogue with broad activity in solid tumors, was performed in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. A total of 26 eligible patients were registered to receive a dose of 1250 mg/m2 weekly for 3 weeks, followed by a 1 week rest. Toxicity was evaluable in 26 patients. Nausea and vomiting occured in 11 and 6 patients, repectively. Grade 3 or 4 hematologic toxicities were infrequent. Two patients developed neutropenic infections. One patient developed fatal liver failure which was thought due to progressive liver metastases or infection 14 days after a single dose of gemcitabine. There were no objective treatment responses (95% CI 0-13%), with a median survival of 6 months in this highly resistant disease population. Gemcitabine is not considered active enough as monotherapy for further evaluation in this disease population.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Antimetabolitos Antineoplásicos/efectos adversos , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Desoxicitidina/efectos adversos , Evaluación de Medicamentos , Femenino , Neoplasias de Cabeza y Cuello/mortalidad , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Ribonucleótido Reductasas/antagonistas & inhibidores , Tasa de Supervivencia , Gemcitabina
2.
Cell Immunol ; 203(2): 84-94, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11006006

RESUMEN

IL-2-activated killer lymphocytes (LAK cells) secrete inflammatory cytokines such as interferon-gamma (IFN-gamma) and tumor necrosis factor alpha (TNFalpha) that can induce nitric oxide (NO) synthesis. We evaluated whether LAK cells could activate NO synthesis in human cancer cells. LAK cells and their culture supernatants induced NO synthesis in DLD-1 colon cancer cells in a dose-dependent manner. NO synthesis was inhibited completely by blocking antibodies to IFN-gamma, demonstrating a key role for this LAK cell cytokine in regulating NO synthesis. The addition of TNFalpha antibodies resulted in partial inhibition. Induction of iNOS mRNA and protein expression in DLD-1 cells was detected. Endogenous NO production inhibited DLD-1 cell proliferation and induced apoptosis, processes that were inhibitable by the NO synthase inhibitor N(G)-monomethyl-l-arginine. Our study has identified a novel, non-contact-dependent LAK cell cytotoxic mechanism: induction of growth inhibition and programmed cell death due to endogenous NO synthesis in susceptible human cancer cells.


Asunto(s)
Apoptosis , Interferón gamma/metabolismo , Células Asesinas Activadas por Linfocinas/metabolismo , Óxido Nítrico/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Antígenos CD/análisis , División Celular , Técnicas de Cocultivo/métodos , Inducción Enzimática , Humanos , Interleucina-2/inmunología , Interleucina-2/farmacología , Células Asesinas Activadas por Linfocinas/citología , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Subgrupos Linfocitarios/clasificación , Subgrupos Linfocitarios/inmunología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Solubilidad , Células Tumorales Cultivadas
3.
Mol Ther ; 2(2): 121-30, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10947939

RESUMEN

Transgene expression and tumor regression after direct injection of plasmid DNA encoding cytokine genes, such as mIL-12 and mIFN-gamma, remain very low. The objective of this study is to develop nontoxic biodegradable polymer-based cytokine gene delivery systems, which should enhance mIL-12 expression, increasing the likelihood of complete tumor elimination. We synthesized poly[alpha-(4-aminobutyl)-l-glycolic acid] (PAGA), a biodegradable nontoxic polymer, by melting condensation. Plasmids used in this study encoded luciferase (pLuc) and murine interleukin-12 (pmIL-12) genes. PAGA/plasmid complexes were prepared at different (+/-) charge ratios and characterized in terms of particle size, zeta potential, osmolality, surface morphology, and cytotoxicity. Polyplexes prepared by complexing PAGA with pmIL-12 as well as pLuc were used for transfection into cultured CT-26 colon adenocarcinoma cells as well as into CT-26 tumor-bearing BALB/c mice. The in vitro and in vivo transfection efficiency was determined by luciferase assay (for pLuc), enzyme-linked immunosorbent assay (for mIL-12, p70, and p40), and reverse transcriptase-polymerase chain reaction (RT-PCR) (for Luc and mIL-12 p35). PAGA condensed and protected plasmids from nuclease degradation. The mean particle size and zeta potential of the polyplexes prepared in 5% (w/v) glucose at 3:1 (+/-) charge ratio were approximately 100 nm and 20 mV, respectively. The surface characterization of polyplexes as determined by atomic force microscopy showed complete condensation of DNA with an ellipsoidal structure in Z direction. The levels of mIL-12 p40, mIL-12 p70, and mIFN-gamma were significantly higher for PAGA/pmIL-12 complexes compared to that of naked pmIL-12. This is in good agreement with RT-PCR data, which showed significant levels of mIL-12 p35 expression. The PAGA/pmIL-12 complexes did not induce any cytotoxicity in CT-26 cells as evidenced by 3-¿4, 5-dimethylthiazol-2-yl¿-2,5-diphenyltetrazolium bromide assay and showed enhanced antitumor activity in vivo compared to naked pmIL-12. PAGA/pmIL-12 complexes are nontoxic and significantly enhance mIL-12 expression at mRNA and protein levels both in vitro and in vivo.


Asunto(s)
Implantes Absorbibles , Citocinas/genética , Citocinas/uso terapéutico , Terapia Genética/métodos , Vectores Genéticos , Neoplasias/terapia , Polímeros , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Glicolatos/síntesis química , Humanos , Interleucina-12/genética , Interleucina-12/uso terapéutico , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Microscopía de Fuerza Atómica , Trasplante de Neoplasias , Plásmidos/genética , Ácido Poliglicólico/análogos & derivados , Ácido Poliglicólico/síntesis química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
5.
Haemophilia ; 5(6): 416-25, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10583529

RESUMEN

The effects of a recombinant factor IX product (BeneFix), and of five plasma-derived factor IX products, AlphaNine, Immunine, Konyne, Mononine and Replinine on in vitro peripheral blood mononuclear cell (PBMC) immune function were compared in a blinded study. We assessed the effects of these products on Con-A-induced lymphocyte proliferation and interleukin-2 and interleukin-10 secretion, expression of lymphocyte activation markers, and nitric oxide secretion by stimulated mouse peritoneal macrophages. At 1 mL-1 for 48 h, Konyne reduced Con-A-induced mitogenesis by 50% (P < 0.05); AlphaNine, Mononine and BeneFix had no effect. At 10 IU mL-1, Con-A-induced mi- togenesis was at control levels with Mononine and BeneFix, but was reduced to <15% (P < 0.05) with each of the other products. IL-2 and IL-10 secretion by Con-A-stimulated lymphocytes was also markedly depressed by all the products tested except Mononine and BeneFix. Dialysis of these products did not substantially affect these results. Flow cytometric analysis of lymphocyte activation markers following Con-A stimulation showed that Konyne also decreased IL-2 receptor alpha and beta chain (CD25 and CD122) induction on PBMC. Konyne also inhibited nitric oxide secretion to levels <18% of controls. These results indicate that certain factor IX products, including some of purported higher purity, substantially depress in vitro immune function. The importance of these findings to in vivo immune function in haemophilia B patients remains to be established.


Asunto(s)
Factor IX/farmacología , Activación de Linfocitos/efectos de los fármacos , Animales , Antígenos de Diferenciación de Linfocitos T/biosíntesis , Antígenos de Diferenciación de Linfocitos T/efectos de los fármacos , Técnicas de Cultivo de Célula , Concanavalina A/farmacología , Citocinas/efectos de los fármacos , Citocinas/metabolismo , Humanos , Terapia de Inmunosupresión , Interferón gamma/farmacología , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Interleucina-4/metabolismo , Lipopolisacáridos/farmacología , Linfocitos/inmunología , Macrófagos/química , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Óxido Nítrico/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Factor de Crecimiento Transformador beta/efectos de los fármacos
6.
Leuk Res ; 23(9): 787-94, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10475617

RESUMEN

The aim of this study is to determine whether the addition of mitoxantrone to high dose cytarabine improves the outcome of treatment in patients with relapsed or refractory acute myeloid leukemia (AML). One hundred and sixty-two eligible patients, 14-76 years of age, with AML either in first relapse or that failed to respond to initial remission induction therapy, with no CNS involvement were randomized to receive therapy with cytarabine 3 gm/M2 i.v. over 2 h every 12 h for 12 doses on days 1-6 (Arm I) (HIDAC); or HIDAC plus mitoxantrone 10 mg/M2 i.v. daily on days 7 9 (Arm II) (HIDAC + M). Patients achieving complete remission were treated with three courses of consolidation including HIDAC (Ara-C 3 gm/M2 i.v. 12 h days 1 3; 2 gm/M2 over age 50) alone (ARM I) or with mitoxantrone (10 mg/M2 i.v. day 1) (ARM II). Among 162 patients (81 HIDAC, 81 HIDAC + M) evaluated for induction toxicity, there were 10 (12%) induction deaths with HIDAC and 13 (17%) with HIDAC + M (2-tailed P = 0.65). Most early deaths were due to infection and/or hemorrhage. Among 162 patients evaluated for responses to induction therapy, 26/81 (32%) HIDAC and 36/81 (44%) HIDAC + M patients achieved complete remission (two-tailed P = 0.15). Although this difference was not statistically significant in univariate analysis, it was after adjusting for the effects of WBC and PMN percentage in multivariate analysis (P=0.013). Median survivals from study entry were 8 months (HIDAC) and 6 months (HIDAC + M); 2-tailed logrank P = 0.58. Among 48 patients registered for consolidation, the median disease-free survivals from that registration were 8 months with HIDAC and 11 months with HIDAC + M (P = 0.60). There were three treatment-related deaths during consolidation (1 HIDAC, 2 HIDAC + M), all due to infections. In this randomized trial, the addition of mitoxantrone to high-dose cytarabine was associated with a trend toward a higher CR rate. There was less evidence for an advantage in disease-free or overall survival, although any such conclusion is limited by the size of the study.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Mieloide/tratamiento farmacológico , Enfermedad Aguda , Adolescente , Adulto , Anciano , Citarabina/administración & dosificación , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mitoxantrona/administración & dosificación , Pronóstico , Recurrencia , Resultado del Tratamiento
7.
Cell Immunol ; 188(1): 33-40, 1998 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-9743555

RESUMEN

We evaluated the effect of antithrombin III (ATIII), a serine protease inhibitor (SERPIN), on induction of nitric oxide (NO) synthesis in murine peritoneal macrophages. Incubation of macrophages with ATIII plus interferon-gamma (IFN-gamma) but not ATIII alone induced nitrite accumulation (a metabolite of NO) in a dose-dependent manner. Expression of the inducible nitric oxide synthase isoform was confirmed by Western blot. NO synthesis was inhibited by NG-monomethyl-l-arginine, by complexing ATIII with thrombin or by rabbit anti-human ATIII antiserum. Addition of polymyxin B to macrophage cultures failed to inhibit ATIII/IFN-gamma-induced NO synthesis, excluding lipopolysaccharide contamination. 125I-ATIII bound to macrophages in a dose-dependent, specific, and saturable manner, with a Km of approximately 7.1 nM. Our results demonstrate that ATIII, but not ATIII/thrombin complex, acts to costimulate macrophage activation and NO synthesis via a novel receptor mediated mechanism, which may indicate a role for SERPINs in macrophage activation.


Asunto(s)
Antitrombina III/farmacología , Macrófagos Peritoneales/inmunología , Óxido Nítrico/biosíntesis , Receptores Inmunológicos/fisiología , Inhibidores de Serina Proteinasa/farmacología , Animales , Antitrombina III/metabolismo , Femenino , Interferón gamma/farmacología , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Activación de Macrófagos/efectos de los fármacos , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Receptores de Superficie Celular/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , omega-N-Metilarginina/farmacología
8.
Transplantation ; 66(4): 413-9, 1998 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-9734481

RESUMEN

BACKGROUND: Previous in vitro studies have suggested that both cytotoxic T lymphocyte (CTL)-mediated and non-CTL-mediated myocyte lysis occur during murine cardiac heterotopic allograft rejection, but the relative importance of these injury mechanisms on myocardial function is not established. We therefore compared the in vivo effects of depletion of CTL and inhibition of nitric oxide synthase (NOS) on contractility of the rejecting heart. METHODS: Syngeneic (BALB/c into BALB/c) and allogeneic (BALB/c into C57/B16) heterotopic abdominal cardiac transplants were performed. In some of the allogeneic transplants, CD8+ lymphocytes were depleted by intraperitoneal injection of anti-CD8 monoclonal antibody. NOS inhibition was accomplished by continuous infusion of NG-monomethyl-L-arginine via a subcutaneous osmotic pump. Five days after transplantation, the abdominal cavity was opened and the transplanted heart exposed. Base to apex developed force was measured during spontaneous beating at a diastolic stretch of 4 g by placing a suture through the apex of the heart and attaching it to a strain gauge. Effects of interventions on graft survival were determined by recording the days required for loss of palpable graft contractions. RESULTS: Allogeneic hearts showed a significant reduction in systolic force compared to non-rejecting syngeneic hearts. Depletion of CD8+ cells improved contractility significantly relative to non-depleted allogeneic hearts, but contractility remained significantly reduced relative to syngeneic hearts. Developed force in allogeneic hearts was also improved by NOS inhibition (P<0.01), and NG-monomethyl-L-arginine infusion slightly prolonged graft survival. CONCLUSION: Both CTL-mediated and NOS-dependent (possibly macrophage-mediated) mechanisms contribute to contractile dysfunction during early cardiac allograft rejection in this model. However, NOS inhibition combined with CTL depletion only slightly prolongs graft survival in this model.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Óxido Nítrico Sintasa/fisiología , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Homólogo
9.
Nitric Oxide ; 2(1): 45-56, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9706742

RESUMEN

Nitric oxide synthesis is strongly induced during IL-2 treatment of mice and humans. While this free radical can act as an antitumor mechanism by inhibiting cellular respiration and DNA synthesis in cancer cells, immunosuppressive effects have also been suggested. We evaluated the effects of NO exposure on the induction of murine lymphokine-activated killer (LAK) cells from splenocytes by IL-2 (6000 IU/ml). When splenocytes were exposed to pure NO gas for 30 min prior to the addition of IL-2, complete abrogation of LAK cell cytotoxicity was observed. In contrast, cytolytic activity of already activated LAK cells was only minimally affected by NO exposure. NO exposure markedly depressed cellular proliferation in response to concanavalin A or IL-2. Immunostaining of LAK cell cultures following NO exposure revealed a marked decrease in CD8+, and peanut lectin (PNA+)/CD56+ subsets (48 and 69%). Dual staining of LAK cells for DNA strand breaks and either PNA or CD8+ identified the induction of programmed cell death in these subsets 12-24 h following NO exposure. These experiments demonstrate that NO has the capacity to inhibit LAK cell induction by inducing apoptosis of cytolytic lymphocyte precursors.


Asunto(s)
Apoptosis , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Óxido Nítrico/farmacología , Animales , Células Cultivadas , Concanavalina A/farmacología , Citotoxicidad Inmunológica , Fragmentación del ADN , Femenino , Inmunohistoquímica , Interleucina-2/inmunología , Interleucina-2/farmacología , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/fisiología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C3H , Nitritos , Bazo/citología , Bazo/inmunología , Células Tumorales Cultivadas
10.
J Immunother ; 21(1): 65-74, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9456439

RESUMEN

Patients treated with high doses of interleukin-2 (IL-2) develop profound anorexia, malaise, loss of energy, mucositis, nausea, and vomiting, which may contribute to poor nutrition. We hypothesized that total parenteral nutrition (TPN) administration would ameliorate these changes and could improve fluid and electrolyte balance. A retrospective analysis of protein and energy intake was performed in 21 sequential patients who received a normal diet (controls) and 16 subsequent patients who received TPN during IL-2 treatment. The effect of TPN on laboratory abnormalities induced by IL-2 was also evaluated. Within 24 h of starting IL-2, mean energy intake declined to 2.5-2.8 kcal/kg in controls in contrast to the energy intake of 25-29 kcal/kg in patients receiving TPN. Protein nutrition was affected in a similar fashion, with a markedly lower protein intake in controls (0.08-0.12 g/kg) than in the TPN group (1.02-1.10 g/kg). TPN improved serum calcium and potassium concentrations, particularly during spontaneous diuresis after completion of IL-2 treatment. Unexpectedly, TPN decreased the frequency and severity of cholestatic jaundice caused by IL-2. Patients receiving TPN had an increased propensity for hyperglycemia and hypophosphatemia. High-dose intravenous bolus IL-2 therapy resulted in a markedly negative nutritional balance in control patients. A brief period of TPN during IL-2 treatment was well tolerated and corrected calorie and protein malnutrition. TPN administration also improved control of serum electrolytes. TPN did not adversely affect tumor progression or patient survival.


Asunto(s)
Carcinoma de Células Renales/terapia , Interleucina-2/administración & dosificación , Interleucina-2/efectos adversos , Neoplasias Renales/terapia , Melanoma/terapia , Nutrición Parenteral Total , Adulto , Nitrógeno de la Urea Sanguínea , Calcio/sangre , Colestasis/etiología , Colestasis/prevención & control , Creatinina/sangre , Proteínas en la Dieta/administración & dosificación , Ingestión de Energía , Femenino , Humanos , Interleucina-2/uso terapéutico , Masculino , Persona de Mediana Edad , Potasio/sangre , Desnutrición Proteico-Calórica/etiología , Desnutrición Proteico-Calórica/prevención & control , Estudios Retrospectivos
11.
Invest New Drugs ; 16(3): 271-4, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10360608

RESUMEN

A phase II trial of Tomudex (raltitrexed, ZD 1694), a new thymidylate synthase inhibitor, was performed in patients with recurrent or metastatic squamous cell carcinoma of the head and neck. This trial demonstrated that Tomudex was well tolerated in this patient population. Nausea and vomiting were minimal, and hematologic toxicities were relatively infrequent. Only one patient was withdrawn from the study due to toxicity (grade 4 diarrhea). One patient exsanguinated from a rent in the carotid artery in an area of tumor involvement, and was categorized as a grade 5 toxicity. Thus 25/27 patients were able to complete at least 2 cycles of treatment. Tomudex demonstrated a 3.7% response rate (95% CI 0.1-19%), with a median survival of 6 months in this highly resistant disease population. Tomudex is not considered active enough as monotherapy for further evaluation in this disease population.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Quinazolinas/uso terapéutico , Tiofenos/uso terapéutico , Anciano , Carcinoma de Células Escamosas/secundario , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Persona de Mediana Edad , Análisis de Supervivencia
13.
J Clin Endocrinol Metab ; 82(5): 1572-81, 1997 May.
Artículo en Inglés | MEDLINE | ID: mdl-9141552

RESUMEN

Recombinant human interleukin-2 (rIL-2) is used to treat refractory cancers. During such treatment, patients develop severe hypocholesterolemia along with striking alterations in the concentration and composition of the circulating lipoproteins. The present study was undertaken to gather information about the pathogenesis of these abnormalities. Patients were studied before-, during- and after a 5-day course of high dose i.v. rIL-2. Whole plasma cholesterol was markedly reduced by rIL-2 administration (52%; P < 0.001), whereas the triglyceride concentration did not change. Thus, the lipoproteins became triglyceride enriched (P = 0.004). Low density lipoprotein cholesterol, apolipoprotein B (apoB), high density lipoprotein cholesterol, and apoA-I concentrations all decreased. Esterified cholesterol levels were markedly reduced. Total plasma apoE increased markedly, and two kinds of abnormal particles appeared: 1) beta-migrating, very low density lipoproteins; and 2) discoidal, apoE- and phospholipid-containing particles with abnormal density and electrophoretic mobility. The activities of two lipoprotein triglyceride hydrolases, lipoprotein lipase and hepatic lipase, fell significantly during treatment and returned promptly to pretreatment levels after rIL-2 was discontinued. Lecithin:cholesteryl acyltransferase (LCAT) activity also decreased significantly (64%) during treatment, but in contrast to the lipases, remained low for at least 5 days after the last dose of rIL-2 (P < 0.001). High dose i.v. rIL-2 induces severe dyslipidemia with deficiencies of both postheparin lipases and acute LCAT deficiency. Most, if not all, of the lipoprotein changes observed are explained by the LCAT deficiency that follows IL-2-induced hepatocellular injury and cholestasis.


Asunto(s)
Interleucina-2/efectos adversos , Deficiencia de la Lecitina Colesterol Aciltransferasa/etiología , Lipasa/deficiencia , Lipoproteína Lipasa/deficiencia , Hígado/enzimología , Apolipoproteína A-I/metabolismo , Apolipoproteínas B/sangre , Apolipoproteínas E/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas , Colesterol/sangre , Ésteres del Colesterol/sangre , Humanos , Interleucina-2/uso terapéutico , Lipoproteínas/sangre , Lipoproteínas HDL/sangre , Lipoproteínas HDL/ultraestructura , Microscopía Electrónica , Neoplasias/tratamiento farmacológico , Fosfolípidos/sangre , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/uso terapéutico , Triglicéridos/sangre
14.
J Immunol ; 155(9): 4382-90, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7594598

RESUMEN

IL-2 therapy is a potent inductive stimulus for nitric oxide (NO.) synthesis in mice and humans. It is not yet clear whether NO. can contribute to IL-2-induced therapeutic responses. The murine skin cancer Meth A is relatively resistant to lymphokine-activated killer (LAK) cell killing, allowing evaluation of the role of IL-2-induced NO. synthesis in vivo, without contribution by LAK cells. Subcutaneous IL-2 treatment of mice bearing i.p. Meth A tumor increased nitrite production by cells derived from ascites (63 +/- 14 microM vs 3.2 +/- 1.5 microM in untreated controls). N omega-monomethyl-L-arginine (MLA), NO. synthase inhibitor, prevented this increase. NO. production correlated in an inverse fashion with tumor cell proliferation in vitro. Evidence for IL-2-induced heme nitrosylation was demonstrated in tumor cells by electron paramagnetic resonance spectroscopy. By immunomagnetic depletion experiments, macrophages were implicated as a major source of NO. synthesis. Cytologic and flow-cytometric evaluation revealed that IL-2 treatment resulted in enhanced lymphocyte and macrophage recruitment into malignant ascites, and decreases in tumor cell recovery. MLA administration further increased host cell recovery. Subcutaneous IL-2 therapy increased urinary nitrate excretion up to eightfold in mice, and appeared to produce a significant survival advantage that was prevented by MLA administration.


Asunto(s)
Ascitis/inmunología , Interleucina-2/uso terapéutico , Óxido Nítrico/biosíntesis , Neoplasias Cutáneas/inmunología , Animales , Arginina/análogos & derivados , Arginina/farmacología , Ascitis/metabolismo , Ascitis/terapia , Femenino , Inmunidad Innata , Células Asesinas Activadas por Linfocinas/inmunología , Activación de Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , NG-Nitroarginina Metil Éster , Óxido Nítrico/inmunología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/farmacología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/terapia , Células Tumorales Cultivadas
15.
J Immunother Emphasis Tumor Immunol ; 18(3): 166-78, 1995 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8770772

RESUMEN

The current study was designed to characterize nitric oxide (NO.) synthesis during interleukin-2 (IL-2) treatment of mice, and to determine whether NO. mediated IL-2-induced "vascular leak." We developed a technique for chronic subcutaneous infusion of the NO. synthase inhibitor N omega monomethyl-L-arginine (MLA) via osmotic minipump to aid in further study of these processes. After IL-2 administration to C3H/HeN mice (180,000 IU i.p. b.i.d. for 5 days), NO. synthesis increased two-to-three fold, peaking on days 5-8. Administration of MLA reduced NO. synthesis in both IL-2-treated mice (from 2.7 to 1 microM/mouse/day), and normal mice (from 1 to 0.5 microM/mouse/day). This agent decreased IL-2-induced radiolabeled albumin accumulation in the liver after i.p. IL-2 administration (p < 0.02). MLA infusions resulted in minimal systemic toxicity in mice, as reflected by complete blood counts or serum chemistries. MLA also did not impair lymphokine-activated killer cell induction in vitro or in vivo, or alter IL-2-induced tumor responses in a 3-day pulmonary metastasis model. These experiments demonstrated that NO. is a mediator involved in the genesis of vascular permeability induced by IL-2 treatment. Studies designed to further evaluate the toxicity and usefulness of MLA infusions to modify this IL-2 induced toxicity appear to be warranted.


Asunto(s)
Interleucina-2/farmacología , Óxido Nítrico/biosíntesis , omega-N-Metilarginina/farmacología , Animales , Permeabilidad Capilar/efectos de los fármacos , Permeabilidad Capilar/fisiología , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/toxicidad , Femenino , Inmunoterapia , Técnicas In Vitro , Bombas de Infusión , Interleucina-2/antagonistas & inhibidores , Interleucina-2/toxicidad , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/inmunología , Masculino , Ratones , Ratones Endogámicos C3H , Óxido Nítrico Sintasa/antagonistas & inhibidores , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/toxicidad , Sarcoma Experimental/inmunología , Sarcoma Experimental/terapia , omega-N-Metilarginina/administración & dosificación , omega-N-Metilarginina/toxicidad
16.
J Immunol ; 152(12): 5796-805, 1994 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8207209

RESUMEN

IL-2 therapy can induce marked oxidative stress via reactive oxygen and nitrogen intermediates. Glutathione, the major intracellular reductant, may become rate limiting to cytotoxic lymphocyte activation and proliferation under these circumstances. N-Acetyl cysteine (NAc-cys) was used to increase intracellular glutathione levels during lymphokine-activated killer (LAK) cell activation by IL-2. Incubation of splenocytes with NAc-cys (0.6 to 1.0 mM) resulted in significant changes in intracellular reduced and total glutathione (92% and 58% increase, respectively) at 96 h. These levels correlated with markedly enhanced cell proliferation (threefold) and cytolytic effector cell generation (> fivefold increase in LU/10(6) cells) induced by the combination of NAc-cys with IL-2. IL-2 exposure by itself unexpectedly increased intracellular reduced glutathione by 43%. IL-2 and NAc-cys were synergistic in increasing glutathione levels (reduced glutathione: 292% increase; total: 251% increase). Inhibition of glutathione synthesis, using L-buthionine-(S,R)-sulfoximine reversed the effects of NAc-cys on intracellular glutathione, as well as cellular proliferation and cytotoxicity. This experiment established that the effects of NAc-cys required de novo glutathione synthesis. In conjunction with IL-2/LAK treatment, oral NAc-cys administration (260 to 900 mg/kg/day for 7 days) significantly decreased tumor progression in a refractory s.c. tumor model. A small fraction of mice (11 to 17%) had complete tumor regressions. NAc-cys may be useful as an adjunct to increase the antitumor activity of IL-2/LAK therapy.


Asunto(s)
Acetilcisteína/uso terapéutico , Fibrosarcoma/terapia , Glutatión/metabolismo , Interleucina-2/uso terapéutico , Acetilcisteína/administración & dosificación , Administración Oral , Animales , Citotoxicidad Inmunológica/efectos de los fármacos , Femenino , Fibrosarcoma/inmunología , Fibrosarcoma/metabolismo , Inmunoterapia , Técnicas In Vitro , Interleucina-2/administración & dosificación , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos/efectos de los fármacos , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C3H , Fenotipo
17.
J Biol Chem ; 269(7): 5127-31, 1994 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-7508933

RESUMEN

The cell-mediated immune response to syngeneic tumors activates the cytokine-inducible nitric oxide synthase. We observed that syngeneic murine tumors exhibited EPR signals related to iron-nitrosyl complex formation. Three different EPR active iron-nitrosyl species were observed, an Fe(RS)2(NO)2 signal and two differentiable heme-nitrosyl complexes. Hemoglobin assays showed that the heme-nitrosyl signals were not derived from contaminating hemoglobin. Signal amplitudes were attenuated in mice treated with N omega-mono-methyl-L-arginine (MLA), an inhibitor of nitric oxide synthase. Tumors grown in vivo contained EPR signals while those grown in culture without continuing cytokine stimulation lost the signals after a few days. Cultured cells that were treated with cytokines, or that were cocultivated with cytokine-activated macrophages, regained EPR active complexes. These results show that the cell-mediated immune response to syngeneic tumors involves the induction of nitric oxide synthase. While nitric oxide synthesis is induced in both tumor infiltrating macrophages and in the tumor cells themselves, only tumor cells contributed to formation of heme-nitrosyl complexes. This result indicates the presence of a novel intracellular target for NO within tumor cells.


Asunto(s)
Arginina/análogos & derivados , Citocinas/farmacología , Espectroscopía de Resonancia por Spin del Electrón/métodos , Hierro/metabolismo , Neoplasias Experimentales/metabolismo , Óxido Nítrico/metabolismo , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Aminoácido Oxidorreductasas/metabolismo , Animales , Arginina/farmacología , Interferón gamma/farmacología , Interleucina-1/farmacología , Hierro/análisis , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Neoplasias Experimentales/química , Óxido Nítrico Sintasa , Células Tumorales Cultivadas/química , Células Tumorales Cultivadas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , omega-N-Metilarginina
18.
Cancer Res ; 53(22): 5507-11, 1993 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-8221691

RESUMEN

The role of macrophages in the host immune response against cancers remains uncertain. Since nitric oxide synthesis represents a significant macrophage antitumor mechanism in vitro, we evaluated whether NO was synthesized during the immune response to growing murine skin cancers. NO synthesis was readily detectable in enzymatically dissociated tumors (RD-995 and LR-298) and was inhibited by N omega-monomethyl-L-arginine (MLA) and by macrophage depletion. Nitrosylation of iron-sulfur and heme complexes was observed in these tumors using electron paramagnetic resonance spectroscopy. NO production in the presence of increasing concentrations of MLA correlated inversely with tumor cell proliferation in vitro. To elucidate the role of NO during in vivo tumor progression, tumor-bearing mice were treated with continuous infusions of the nitric oxide synthase inhibitor MLA. MLA-treated mice demonstrated increased growth and delayed rejection of the highly antigenic UV radiation-induced regressor tumor LR-298. These experiments demonstrate that macrophage-derived NO synthesis can contribute to the antitumor immune response in vivo.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Macrófagos/metabolismo , Neoplasias Inducidas por Radiación/inmunología , Óxido Nítrico/biosíntesis , Neoplasias Cutáneas/inmunología , Animales , Arginina/administración & dosificación , Arginina/farmacología , Carcinoma de Células Escamosas/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Rechazo de Injerto , Macrófagos/fisiología , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Neoplasias Inducidas por Radiación/metabolismo , Óxido Nítrico/inmunología , Neoplasias Cutáneas/metabolismo , Organismos Libres de Patógenos Específicos , Células Tumorales Cultivadas , Rayos Ultravioleta
19.
J Immunother Emphasis Tumor Immunol ; 13(1): 49-54, 1993 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8435432

RESUMEN

A patient with interleukin (IL)-2 responsive metastatic melanoma developed meningeal carcinomatosis. Treatment was attempted with intrathecal (i.t.) IL-2 (5 weekly doses of 3-6 x 10(6) IU) without glucocorticosteroids. Marked increases in cerebrospinal fluid (CSF) pressure occurred 5-10 h following each IL-2 dose, resulting in reversible abnormalities of neurologic function. IL-2 clearance from the CSF ranged from 21 to 85 ml/h, with an apparent first order rate constant of 0.08-0.23 hr-1. These values were consistent with clearance by bulk flow mechanisms. Clearance also correlated directly with peak CSF pressure. Progressive increases in CSF tumor necrosis factor (TNF)-alpha and IL-6 levels, but not Il-1 alpha, were also noted over successive treatment cycles. Increasing neutrophilia (peaking at 12 h postdose) and a delayed lymphocytosis and monocytosis (at 20-30 h) were observed with each successive i.t. IL-2 dose. Activated lymphocytes were not observed in the CSF, however, suggesting that an exogenous source of activated lymphokine-activated killer (LAK) cells may be helpful in obtaining effective antitumor responses.


Asunto(s)
Carcinoma/secundario , Interleucina-2/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Meníngeas/secundario , Adulto , Carcinoma/tratamiento farmacológico , Líquido Cefalorraquídeo/citología , Presión del Líquido Cefalorraquídeo/efectos de los fármacos , Femenino , Humanos , Interleucina-2/efectos adversos , Interleucina-2/farmacocinética , Interleucina-6/líquido cefalorraquídeo , Presión Intracraneal/efectos de los fármacos , Células Asesinas Activadas por Linfocinas , Neoplasias Meníngeas/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/líquido cefalorraquídeo
20.
J Androl ; 13(5): 437-43, 1992.
Artículo en Inglés | MEDLINE | ID: mdl-1331012

RESUMEN

It was recently observed that treatment of patients with a high dosage of human interleukin (IL-2) resulted in suppression of plasma concentrations of testosterone. A murine model was developed to assess the direct and indirect effects of murine IL-2 and the secondarily released cytokines, gamma interferon (INF gamma), and tumor necrosis factor (TNF alpha), on testosterone production in isolated Leydig cells. Pretreatment for 24 hours with IL-2 (100 to 500 IU/ml) or INF gamma (100 to 1000 IU/ml) significantly decreased testosterone production in response to luteinizing hormone (LH; P < 0.02 and 0.005, respectively). The combinations of INF gamma with either TNF alpha or IL-2 produced enhanced suppressive effects on Leydig cell testosterone production. Steroidogenic precursors (22-hydroxycholesterol, 17 alpha-hydroxypregnenolone, and dehydroepiandrosterone) restored testosterone secretion to control levels after preincubation with INF gamma or TNF alpha. In contrast, the inhibition of testosterone synthesis produced by either IL-2 or INF gamma plus TNF alpha could be reversed by 17 alpha-hydroxypregnenolone and dehydroepiandrosterone, but not by 22-hydroxycholesterol (P < 0.01). Dibutyryl cyclic adenosine monophosphate was also ineffective in reversing the inhibitory effects of these cytokines on synthesis. Although IL-2 directly inhibited synthesis in isolated Leydig cells, it stimulated testosterone production (P < 0.005) in minced murine testes. This suggests that IL-2 releases regulatory factors from other cells that were able to overcome the direct inhibitory effect of IL-2. This stimulatory effect was not caused by INF gamma and TNF alpha because INF gamma alone or with TNF alpha inhibited (P < 0.005) testosterone production in minced testes.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Interferón gamma/farmacología , Interleucina-2/farmacología , Células Intersticiales del Testículo/metabolismo , Testosterona/biosíntesis , Factor de Necrosis Tumoral alfa/farmacología , Animales , Bucladesina/farmacología , Técnicas In Vitro , Células Intersticiales del Testículo/efectos de los fármacos , Hormona Luteinizante/farmacología , Masculino , Ratones , Testículo/efectos de los fármacos , Testículo/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA