Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cancer Gene Ther ; 14(8): 717-23, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17557109

RESUMEN

Effective eradication of established tumor and generation of a lasting systemic immune response are the goals of cancer immunotherapy. The objective of this phase IB study was to assess the safety and toxicity of treatment to metastatic tumor underlying the skin with the DNA encoding interleukin-12 (IL-12). This treatment strategy allowed the patient's own tumor to serve as a source of autologous antigen in the tumor microenvironment. We proposed that IL-12 protein produced by the transfected cells would result in the generation of both a local and systemic antitumor response. The tumor was treated with either three or six intratumoral injections of plasmid containing IL-12 DNA. This treatment strategy resulted in no significant local or systemic toxicity. The treatment did not result in an increase in serum IL-12 protein. The size of the treated lesion decreased significantly (greater than 30%) in five of the 12 patients. However, nontreated subcutaneous lesions or other disease did not decrease in size.


Asunto(s)
ADN de Neoplasias/administración & dosificación , Vectores Genéticos/administración & dosificación , Interleucina-12/administración & dosificación , Interleucina-12/genética , Melanoma/terapia , Plásmidos/genética , Adulto , Anciano , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/terapia , ADN de Neoplasias/efectos adversos , Femenino , Vectores Genéticos/efectos adversos , Humanos , Inyecciones Intralesiones , Interleucina-12/efectos adversos , Neoplasias Renales/genética , Neoplasias Renales/terapia , Masculino , Melanoma/genética , Melanoma/secundario , Persona de Mediana Edad
2.
Clin Cancer Res ; 7(4): 952-61, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11309346

RESUMEN

Particle-mediated gene delivery was used to immunize mice against melanoma. Mice were immunized with a plasmid cDNA coding for the human melanoma-associated antigen, gp100. Murine B16 melanoma, stably transfected with human gp100 expression plasmid, was used as a tumor model. Particle-mediated delivery of gp100 plasmid into the skin of naïve mice resulted in significant protection from a subsequent tumor challenge. Co-delivery of murine granulocyte-macrophage colony-stimulating factor (GM-CSF) expression plasmid together with the gp100 plasmid consistently resulted in a greater level of protection from tumor challenge. The inclusion of the GM-CSF plasmid with the gp100 DNA vaccine allowed a reduction in the gp100 plasmid dose required for antitumor efficacy. Protection from tumor challenge was achieved with as little as 62.5 ng of gp100 DNA per vaccination. Tumor protection induced by the gp100 + GM-CSF gene combination was T cell mediated, because it was abrogated in vaccinated mice treated with anti-CD4 and anti-CD8 monoclonal antibodies. In addition, administration of the gp100 + GM-CSF DNA vaccine to mice bearing established 7-day tumors resulted in significant suppression of tumor growth. These results indicate that inclusion of GM-CSF DNA augments the efficacy of particle-mediated vaccination with gp100 DNA, and this form of combined gp100 + GM-CSF DNA vaccine warrants clinical evaluation in melanoma patients.


Asunto(s)
ADN/uso terapéutico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Melanoma Experimental/prevención & control , Glicoproteínas de Membrana/uso terapéutico , Proteínas de Neoplasias/uso terapéutico , Animales , ADN/administración & dosificación , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma Experimental/tratamiento farmacológico , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/genética , Plásmidos/genética , Recurrencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Vacunación , Antígeno gp100 del Melanoma
3.
Clin Cancer Res ; 3(8): 1277-88, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9815810

RESUMEN

We conducted a Phase IB trial of antidisialoganglioside chimeric 14. 18 (ch14.18) antibody and interleukin 2 (IL-2) to determine the maximal tolerated dose (MTD), immunological effects, antitumor effects, and toxicity of this treatment combination. Twenty-four melanoma patients received immunotherapy with ch14.18 antibody and a continuous infusion of Roche IL-2 (1.5 x 10(6) units/m2/day) given 4 days/week for 3 weeks. The ch14.18 antibody (dose level, 2-10 mg/m2/day) was scheduled to be given for 5 days, before, during, or following initial systemic IL-2 treatment. The ch14.18 MTD was 7.5 mg/m2/day, and 15 patients were treated with the ch14.18 MTD. Immunological effects included the induction of lymphokine-activated killer activity and antibody-dependent cellular cytotoxicity by peripheral blood mononuclear cells. In addition, serum samples obtained following ch14.18 infusions were able to facilitate in vitro antibody-dependent cellular cytotoxicity. Antitumor activity included one complete response, one partial response, eight patients with stable disease, and one patient with >50% decrease of hepatic metastases in the face of recurrence of a s.c. lesion. Dose-limiting toxicities were a severe allergic reaction and weakness, pericardial effusion, and decreased performance status. Most patients treated at the MTD had abdominal, chest, or extremity pain requiring i.v. morphine. One patient had an objective peripheral neuropathy. This IL-2 and ch14.18 treatment combination induces immune activation in all patients and antitumor activity in some melanoma patients. We are attempting to enhance this treatment approach by addition of the anti-GD3 R24 antibody to this IL-2 and ch14.18 regimen.


Asunto(s)
Anticuerpos Monoclonales/efectos adversos , Interleucina-2/efectos adversos , Melanoma/terapia , Adulto , Anticuerpos Antiidiotipos/sangre , Citotoxicidad Celular Dependiente de Anticuerpos , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Inmunofenotipificación , Inmunoterapia , Células Asesinas Activadas por Linfocinas/inmunología , Recuento de Linfocitos , Linfocitos/inmunología , Masculino , Melanoma/inmunología , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes/efectos adversos , Células Tumorales Cultivadas
4.
Clin Cancer Res ; 2(2): 319-30, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9816175

RESUMEN

Interleukin 2 (IL-2) and granulocytes-macrophage colony-stimulating factor (GM-CSF) are activators of the lymphocyte and granulocyte/macrophage series, respectively. We conducted a phase IB trial to identify the maximally tolerated dose and to assess immunological effects of the combination. Thirty-four patients with incurable cancers received 2.5, 5, or 10 microgram/kg GM-CSF s.c. either before or concurrently with 1.5 or 3.0 million units/m2/day IL-2. The most common laboratory and clinical side effects included an elevation of the total WBC or eosinophil count due to GM-CSF, and constitutional symptoms due to IL-2. Grade 3 or 4 toxicities included hypotension, thrombocytopenia, elevations in aspartate aminotransferase or bilirubin, renal toxicity, gastrointestinal hemorrhage, arrhythmia, and constitutional symptoms. Two patients receiving 5.0 microgram/kg GM-CSF plus concurrent 3.0 million units IL-2 experienced dose-limiting grade 3 or 4 neurological toxicity, which reversed almost completely. An increase in the serum-soluble IL-2 alpha chain receptor was observed with administration of GM-CSF, IL-2, or the combination. IL-2 therapy enhanced lymphokine-activated killer activity, antibody-dependent cellular cytotoxicity, and lymphocyte activation, with increased CD16 and CD56 expression. GM-CSF increased expression of human leukocyte antigen DR on peripheral blood monocytes and decreased surface expression of CD16 on circulating monocytes and polymorphonuclear cells. Lymphokine-activated killer activity and CD16 expression on monocytes and lymphocytes and CD56 expression on lymphocytes were significantly lower in patients receiving GM-CSF simultaneously with IL-2 than in patients receiving the sequential treatment. Antitumor activity was observed in the lungs of four of eight renal cell carcinoma patients with pulmonary metastases treated with concurrent GM-CSF and IL-2. Although no or minimal shrinkage was observed in the patients' large primary tumors, these results warrant further study. The recommended initial Phase II dose and schedule is 1.25 microgram/kg/day GM-CSF, given concurrently with 1.5 million Roche units/m2/day (4.5 x 10(6) international units/m2/day) IL-2, with subsequent escalation of GM-CSF to 2.5 microgram/kg/day after careful observation for toxicities.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Interleucina-2/administración & dosificación , Neoplasias/terapia , Adulto , Anciano , Citotoxicidad Celular Dependiente de Anticuerpos , Antígeno CD56/análisis , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Antígenos HLA-DR/análisis , Humanos , Interleucina-2/efectos adversos , Células Asesinas Activadas por Linfocinas/inmunología , Masculino , Persona de Mediana Edad , Neoplasias/inmunología , Receptores de IgG/análisis , Receptores de Interleucina-2/análisis
5.
Hum Gene Ther ; 8(7): 875-91, 1997 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-9143914

RESUMEN

The objective of this Phase I study is to assess the acute and long-term toxicities of intradermal vaccination of cancer patients with lethally-irradiated tumor cells that have been transfected by particle-mediated gene transfer (PMGT) with gold particles coated with human granulocyte-macrophage colony stimulating factor (GM-CSF) DNA in a plasmid expression vector. The GM-CSF DNA-coated gold particles are delivered to tumor cells using helium pressure with a hand held gene delivery device. Preclinical studies have demonstrated that vaccination of mice with irradiated, GM-CSF-transfected melanoma cells provided protection from subsequent challenges with non-irradiated, non-transfected tumor cells. Ongoing human tumor immunotherapy studies use patients' melanoma or renal carcinoma cells transfected with a retroviral vector containing GM-CSF cDNA as a vaccine to elicit anti-tumor immune responses. PMGT transfection, unlike retroviral transfection, does not require tumor cells to proliferate in vitro to undergo gene transfer. Instead, tumor tissue can be dissociated into small tissue clumps or cell aggregates and then immediately transfected using the gene gun. PMGT physically inserts the DNA without the need for cell surface interaction with viral components or exposure of the patient to viral antigens. As described in this protocol, fresh human sarcoma and melanoma specimens can be transfected with the GM-CSF DNA-coated gold particles with subsequent production of biologically active GM-CSF protein. In this study tumor tissue will be obtained from patients with melanoma or sarcoma. Tumor tissue will be dissociated, irradiated, and transfected with GM-CSF DNA by PMGT. In this ascending dosage study, two dose levels of GM-CSF DNA will be studied in 2 groups of 6 patients each. Patients will receive two intradermal injections of the irradiated, transfected tumor in a single extremity. On days 3 and 14 post-vaccination, patients will undergo surgical excision of the vaccination sites to assess GM-CSF production and infiltration of immune effector cells. On Day 25, patients will undergo DTH testing with intradermal injection in their opposite extremity of 5 x 10(6) irradiated non-transfected autologous tumor cells cryopreserved at the time of vaccine preparation. This injection site will be assessed on day 28 post-vaccination and surgical excision of the DTH testing site will be performed on day 28 if a positive reaction is noted. The patients will be observed for local and systemic toxicity on days 2, 3, 5, 8, 14, 25, and 28 after the vaccination. Restaging of the patients' disease and long term toxicity evaluation will be performed at 3, 6, and 12 months and then yearly.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Técnicas de Transferencia de Gen , Terapia Genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/terapia , Sarcoma/terapia , Animales , Vacunas contra el Cáncer/efectos adversos , Protocolos Clínicos , Ensayos Clínicos Fase I como Asunto , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos adversos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Melanoma/inmunología , Ratones , Sarcoma/inmunología , Transfección , Células Tumorales Cultivadas
6.
Hum Gene Ther ; 9(13): 1851-61, 1998 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9741424

RESUMEN

Cytokine gene-engineered tumor vaccines are currently an area of intense investigation in both basic research and clinical medicine. Our efforts to utilize tumor vaccines in an immunotherapeutic manner involve canines with spontaneous tumors. We hypothesized that canine tumor cells, transfected with human granulocyte-macrophage colony-stimulating factor (hGM-CSF) cDNA in a plasmid vector, would prove nontoxic following intradermal administration, generate biologically relevant levels of protein, effect local histological changes at the sites of vaccination, and create a systemic antitumor response. Sixteen tumor-bearing dogs were admitted to a study of ex vivo gene therapy. Tumor tissue was surgically removed, enzymatically and mechanically dissociated, irradiated, transfected, and intradermally injected back into the patients. The dogs were vaccinated with primary autologous tumor cells transfected with hGM-CSF or a reporter control gene. hGM-CSF protein was detected (0.07 to 14.15 ng/vaccination site) at 24 hr postinjection and dramatic histological changes were observed, characterized by neutrophil and macrophage infiltration at the sites of injection of hGM-CSF-transfected tumor cells. This was in stark contrast to the lesser neutrophilic and eosinophilic infiltrates found at control vaccination sites. Objective evidence of an antitumor response was observed in three animals. These data, in a large animal translational model of spontaneous tumors, demonstrate in vivo biological activity of hGM-CSF-transfected autologous tumor cell vaccines.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Enfermedades de los Perros/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias/veterinaria , Animales , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Trasplante de Células/veterinaria , ADN Complementario , Enfermedades de los Perros/patología , Perros , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Humanos , Masculino , Trasplante de Neoplasias/veterinaria , Neoplasias/patología , Neoplasias/terapia , Transfección/métodos , Células Tumorales Cultivadas , Vacunación/veterinaria
7.
J Immunother (1991) ; 11(1): 30-9, 1992 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-1734946

RESUMEN

The design of combination hormonal and immunotherapeutic protocols for breast cancer patients may be facilitated by analysis of preclinical in vitro model systems. Estrogen receptor positive (ER+: MCF-7) and negative (ER-: MDA-MB-231) human breast cancer cell lines were utilized to evaluate the effects of tamoxifen (TAM) and estradiol (E2) on modulation of breast cancer target susceptibility to lysis by lymphokine-activated killer (LAK) cells. E2-stimulated ER+ cells were more susceptible to lysis by LAK cells than corresponding TAM-treated or control cells, while treatment of ER- cells with either E2 or TAM alone did not alter from control their susceptibility to this immune-mediated lysis. All ER+ and ER- cells tested remained sensitive after treatment with TAM to lysis by LAK cells. In addition, an adenocarcinoma reactive human-mouse chimeric monoclonal antibody (ING-1) was able to significantly boost in vivo generated LAK cell-mediated lysis of control, E2-treated, and TAM-treated ER+ and ER- cells. These in vitro results provide a preclinical rationale for in vivo testing of TAM, interleukin-2 (IL-2), and breast cancer reactive antibody-dependent cellular cytotoxicity facilitating antibody in patients with refractory or high risk breast cancer.


Asunto(s)
Neoplasias de la Mama/inmunología , Estradiol/farmacología , Células Asesinas Activadas por Linfocinas/inmunología , Tamoxifeno/farmacología , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Ciclo Celular , Terapia Combinada , Humanos , Interleucina-2/farmacología , Modelos Biológicos , Receptores de Estrógenos/análisis , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/inmunología
8.
Cancer Gene Ther ; 8(2): 118-27, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11263527

RESUMEN

IL-12 gene therapy results in tumor regression in some, but not all, murine models. We hypothesized that expression of B7.1 on the tumor cell surface was necessary for IL-12-mediated tumor regression. In addition, we hypothesized that all cells must express B7.1 for this to be effective. To evaluate this hypothesis, tumor nodules were established in mice with either wild-type B16 melanoma or with B16 melanoma modified to express B7.1. IL-12 cDNA was transferred to the tumor by particle-mediated gene transfer. All tumors modified to express B7.1 regressed completely after IL-12 cDNA treatment. When the percent of B7.1-transfected B16 cells was decreased to 50%, no animals survived after treatment. Animals rendered tumor-free were then challenged with wild-type B16. Fifty percent of mice was protected from this tumor challenge. Expression of CD28 (the stimulatory B7.1 ligand) was significantly increased in both CD8(+) T cells and natural killer cell populations of mice rejecting tumor challenge compared to mice with tumor growth. These results suggest that the costimulatory molecule B7.1 is required for initial tumor sensitivity to IL-12 gene therapy and that protection from subsequent challenge with B7.1 (-) tumor is mediated by CD28(+) immune effector cells.


Asunto(s)
Antígeno B7-1/metabolismo , Terapia Genética , Inmunoconjugados , Interleucina-12/genética , Melanoma Experimental/terapia , Abatacept , Animales , Antígenos CD , Antígenos de Diferenciación/metabolismo , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/metabolismo , Antígeno CTLA-4 , Femenino , Citometría de Flujo , Células Asesinas Naturales/metabolismo , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos C57BL , Bazo/patología , Transfección
9.
Cancer Gene Ther ; 3(3): 192-201, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8725884

RESUMEN

The aim of this study was to determine if human melanoma cells could be molecularly modified by particle-mediated gene transfer with a "gene gun", using genes for interferon-gamma (IFN-gamma), the B7-1 costimulatory molecule (CD80), and human leukocyte antigen (HLA)-A2, to augment expression of both HLA molecules and B7-1. Established and early passage melanoma cells transfected with human IFN-gamma complementary DNA (cDNA) produced IFN-gamma (50-5,000 pg/mL). The biological effect of this IFN-gamma transgene included an upregulation, or de novo appearance, of HLA expression. These melanoma cells had no detectable baseline surface expression of the B7-1 costimulatory molecule, but 8% to 31% of these cells became B7-1 positive with no selection procedure after gene transfer with human B7-1 cDNA. After combination gene transfer with cDNAs for both IFN-gamma and B7-1, 9% to 33% of these cells expressed both HLA-DR and B7-1. In combination gene transfer experiments with cDNAs for both HLA-A2 and B7-1, dual expression of HLA-A2 and B7-1 was achieved in 10% to 17% of the melanoma cells. Thus, the molecular modification of human melanoma cells to increase expression of both HLA and B7-1 can be achieved by particle-mediated gene delivery and presents a promising strategy to stimulate antimelanoma T-cell immunity. Key words: Melanoma; T cells; B7-1 costimulatory molecule (CD80); major histocompatibility complex.


Asunto(s)
Antígeno B7-1/biosíntesis , Antígenos HLA/biosíntesis , Interferón gamma/biosíntesis , Melanoma , Transfección/métodos , Antígeno B7-1/genética , Biolística , Línea Celular , ADN Complementario , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Expresión Génica , Genes Reporteros , Terapia Genética/métodos , Antígeno HLA-A2/biosíntesis , Antígenos HLA-DR/biosíntesis , Humanos , Interferón gamma/genética , Complejo Mayor de Histocompatibilidad , Melanoma/terapia , Plásmidos , Proteínas Recombinantes/biosíntesis , Células Tumorales Cultivadas , beta-Galactosidasa/biosíntesis
10.
Cancer Gene Ther ; 6(1): 26-36, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10078961

RESUMEN

Tumor vaccines and gene therapy have received significant attention as means of increasing cellular and humoral immune responses to cancer. We conducted a pilot study of seven research dogs to determine whether intradermal injection of canine tumor cells transfected via the Accell particle-mediated gene transfer device with the cDNA for human granulocyte-macrophage colony-stimulating factor (hGM-CSF) would generate biologically relevant levels of protein and result in demonstrable histological changes at sites of vaccination. Tumor cell vaccines of 10(7) irradiated canine melanoma cells were nontoxic, safe, and well tolerated. No significant alterations in blood chemistry values or hematological profiles were detected. A histological review of control vaccine sites revealed inflammatory responses predominated by eosinophils, whereas vaccine sites with hGM-CSF-transfected tumor cells had an influx of neutrophils and macrophages. Enzyme-linked immunosorbent assays of skin biopsies from vaccine sites had local hGM-CSF production (8.68-16.82 ng/site of injection) at 24 hours after injection and detectable levels (0.014-0.081 ng/site) for < or =2 weeks following vaccination. Flow cytometric analysis of hGM-CSF-transfected cells demonstrated < or =25% transfection efficiency, and hGM-CSF levels obtained during time-course assays demonstrated biologically relevant levels for both irradiated and nonirradiated samples. These data demonstrate the in vivo biological activity of irradiated hGM-CSF-transfected canine tumor cells and help provide evidence for a valid translational research model of spontaneous tumors.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Técnicas de Transferencia de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma/genética , Animales , Muerte Celular , Perros , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Ensayo de Inmunoadsorción Enzimática , Eosinófilos/inmunología , Genes Reporteros , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Luciferasas/metabolismo , Proyectos Piloto , Transfección , Células Tumorales Cultivadas , beta-Galactosidasa/metabolismo
11.
Cancer Lett ; 111(1-2): 111-5, 1997 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9022135

RESUMEN

It has been proposed that immunoregulatory cytokines play a role in the onset and development of cancer cachexia, although evidence supporting this theory remains inconclusive. In the present study, SCID mice were implanted with one of two tumor cell lines known to induce weight loss in rats. Growth of the Morris 7777 hepatoma was associated with weight loss as well as increased levels of tumor necrosis factor and interleukins 1 and 6 in spleen cells of tumor-bearing mice. Growth of the MCA sarcoma did not induce weight loss, nor did it increase cytokine expression in spleen cells of tumor-bearing mice. We conclude that increased cytokine expression is associated with weight loss in tumor-bearing SCID mice, and immune activation for cytokine expression does not require the presence of T or B cells.


Asunto(s)
Interleucina-1/metabolismo , Interleucina-6/metabolismo , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/fisiopatología , Proteínas de Neoplasias/metabolismo , Sarcoma Experimental/metabolismo , Sarcoma Experimental/fisiopatología , Factor de Necrosis Tumoral alfa/metabolismo , Pérdida de Peso/fisiología , Animales , Neoplasias Hepáticas Experimentales/patología , Masculino , Ratones , Ratones SCID , Ratas , Ratas Endogámicas BUF , Sarcoma Experimental/patología , Especificidad de la Especie
12.
Mutat Res ; 476(1-2): 83-97, 2001 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-11336986

RESUMEN

T-cell activation by malignant melanoma would be anticipated to stimulate T-cell proliferation, which in turn has been associated with increasing the likelihood of somatic gene mutation. The purpose of this study was to test the hypothesis that in vivo hypoxanthine guanine phosphoribosyltransferase (hprt) mutant frequencies (MFs) are increased in peripheral blood T-cells from melanoma patients compared to normal controls. Assays were made of 48 peripheral blood samples from melanoma patients with stage 3 (13 patients) and stage 4 (35 patients) disease, 38 normal controls, and of nine tumor bearing lymph nodes. The mean hprt log(10)(MF) in patient peripheral blood was -4.77 (geometric mean hprt MF=17.0x10(-6)) compared to a mean hprt log(10)(MF) of -4.87 (geometric mean hprt MF=13.5x10(-6)) in controls. Although modest, this difference is statistically significant both by t-test (P=0.049) and after adjustment for covariates of age, gender, and cigarette smoking by regression analysis (P=0.001). Among the melanoma patients, the mean log(10)(MF) for the 17 patients who had received potentially genotoxic therapies was not significantly different from the mean log(10)(MF) for the 31 patients not receiving such therapies. The hprt MFs in the nine tumor bearing nodes were compared with MFs in peripheral blood from the same patients and revealed a non-significant (P=0.07) trend for increasing MFs in blood. Furthermore, analyses of T-cell receptor gene rearrangement patterns revealed hprt mutants originating from the same in vivo clone in both peripheral blood and a tumor-bearing node. The finding of elevated hprt MFs not entirely explained by genotoxic therapies in patients compared to controls can be explained either by hypermutability or in vivo T-cell activation. The similar MFs in peripheral blood and tumor bearing lymph nodes, as well as the finding of mutant representatives of the same in vivo T-cell clone in both locations, support monitoring peripheral blood to detect events in the nodes. If in vivo proliferation accounts for the current findings, the hprt deficient (hprt-) mutant fraction in blood may be enriched for T-cells that mediate the host immune response against malignant melanoma. Further studies will characterize the functional reactivity of hprt mutant isolates against melanoma-related antigens.


Asunto(s)
Melanoma/genética , Melanoma/inmunología , Mutación , Linfocitos T/inmunología , Tioguanina/toxicidad , Adulto , Anciano , Estudios de Casos y Controles , Resistencia a Medicamentos/genética , Femenino , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Técnicas In Vitro , Metástasis Linfática , Activación de Linfocitos , Masculino , Melanoma/enzimología , Melanoma/secundario , Persona de Mediana Edad , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología
13.
Med Oncol ; 17(1): 64-9, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10713663

RESUMEN

An association between auto-immune disorders and interferon (IFN) has been reported. High levels of natural IFNalpha are present in the blood of patients with auto-immune disease and correlate with disease activity. In addition, IFNalpha treatment of humans has resulted in multiple reports of associated auto-immune phenomena. We describe a patient who underwent resection of regionally metastatic melanoma, was given adjuvant high-dose IFNalpha2b, and subsequently developed dermatomyositis. To the authors' knowledge this is the first report of dermatomyositis in association with IFNalpha treatment. We review the literature reporting associations between IFNalpha and auto-immune disease and discuss possible mechanisms by which IFNalpha may contribute to the development of auto-immune disease. High dose IFNalpha2b is more commonly prescribed since it was approved as an adjuvant treatment for patients with surgically resected high-risk melanoma. The potential for cases of IFN-associated auto-immune disease is therefore a clinical concern. Standard side effects of high-dose IFN therapy resemble symptoms of auto-immune diseases, which may make prompt diagnosis difficult. Therefore, it is important that auto-immune diseases such as dermatomyositis are recognized as potential side effects of treatment with high-dose IFNalpha.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Dermatomiositis/inducido químicamente , Interferón-alfa/efectos adversos , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Enfermedades Autoinmunes/diagnóstico , Dermatomiositis/diagnóstico , Dermatomiositis/patología , Diagnóstico Diferencial , Femenino , Humanos , Interferón-alfa/uso terapéutico , Melanoma/patología , Persona de Mediana Edad , Neoplasias Cutáneas/patología
15.
J Immunother Emphasis Tumor Immunol ; 14(4): 329-35, 1993 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-8280716

RESUMEN

Recent technical advances have enabled the generation of clinical reagents for immunotherapy. Currently, treatment protocols combining both interleukin-2 (IL-2) and tumor-specific monoclonal antibody are underway at the University of Wisconsin Comprehensive Cancer Center and elsewhere. These approaches are based on the hypothesis that IL-2-activated lymphocytes will use tumor-reactive antibody to more selectively and effectively destroy tumor in vivo. Just as IL-2 can activate lymphocytes to destroy antibody-coated tumor cells, other agents can activate neutrophils and monocytes to destroy antibody-treated tumor cells. We are investigating, in laboratory and clinic, approaches aimed at eventually using combinations of distinct antibody-based tumor recognition mechanisms in patients whose monocytes, neutrophils, and lymphocytes have been simultaneously activated with multiple biologic agents.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Interleucina-2/uso terapéutico , Neoplasias/terapia , Animales , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Humanos , Inmunoterapia , Neoplasias/inmunología , Tumor de Wilms/terapia
16.
Cancer Immunol Immunother ; 49(2): 85-93, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10823418

RESUMEN

The aim of this study was to compare the antigenicity of human melanoma cells molecularly modified by particle-mediated gene transfer to have transient or stable expression of the B7-1 co-stimulatory molecule (CD80). The unmodified melanoma cells (mel5, m21) had no constitutive expression of B7-1, but 22%-28% of cells had transient B7-1 expression 24 h following transfection with cDNA for B7-1 (mel5-B7, m21-B7). In addition, 85%-90% of cells had stable B7-1 expression following transfection with cDNA for B7-1 and in vitro culture under selection conditions (mel5-B7neo, m21-B7neo). Allogeneic HLA-unmatched normal donor peripheral blood mononuclear cells (PBMC) secreted greater amounts of granulocyte/macrophage-colony-stimulating factor (GM-CSF) when incubated for 3 days with m21-B7neo than did PBMC incubated with m21-B7, which, in turn, secreted greater amount of GM-CSF than PBMC incubated with m21. Similarly, cell-mediated cytotoxicity against unmodified melanoma cells by PBMC co-cultured for 5 days with the modified or unmodified melanoma cells was proportional to the level of B7-1 expression on the stimulating cells. This cytolytic activity had both an HLA-class-I-restricted and an HLA-class-I-unrestricted component. Following 5 days of co-culture, PBMC expression of CD28, the ligand for B7-1, was down-regulated in proportion to the level of B7-1 expression on the stimulating melanoma cells. Thus, particle-mediated gene delivery of cDNA for B7-1 into human melanoma cells increased expression of functional B7-1 and enhanced the antigenicity of the gene-modified cells in proportion to their level of B7-1 expression.


Asunto(s)
Antígeno B7-1/fisiología , Melanoma/inmunología , Antígeno B7-1/genética , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Antígenos HLA-A/genética , Humanos , Inmunofenotipificación , Activación de Linfocitos , Transfección , Células Tumorales Cultivadas
17.
Cancer ; 75(11): 2737-41, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7743479

RESUMEN

BACKGROUND: Splenomegaly in patients with cancer raises the suspicion of tumor involvement. Splenic enlargement in the absence of splenic metastases, however, has been reported in patients treated with interleukin-2 (IL-2) immunotherapy. This study characterizes the change in spleen size that occurred in 42 patients treated with IL-2 between 1989 and 1993 for nonhematologic malignancies. METHODS: Computed tomography (CT) scans before and during immunotherapy were available for review on all 42 patients and after immunotherapy on 16 of these patients. The splenic index was measured for each CT by a single reader blinded to the time course of IL-2 therapy. RESULTS: Mean splenic index increased 64.1% from 646 cm3 (standard deviation [SD], 387) pre-IL-2 to 1059 cm3 (SD, 534) during therapy with IL-2 (P < 0.0001). The splenic index remained elevated at 1112 cm3 (SD, 633) after completion of IL-2 therapy. CONCLUSIONS: Splenomegaly, temporally associated with IL-2 therapy for nonhematologic malignancies, is likely to represent a sequela of therapy and not tumor progression.


Asunto(s)
Interleucina-2/efectos adversos , Esplenomegalia/etiología , Adulto , Anciano , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunoterapia/efectos adversos , Masculino , Persona de Mediana Edad , Esplenomegalia/patología
18.
J Biol Response Mod ; 9(5): 456-62, 1990 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2254758

RESUMEN

Eleven patients receiving weekly cycles of therapy with recombinant interleukin-2 (IL-2) were evaluated with a sensitive limiting dilution analysis to determine lymphokine-activated killer (LAK) cell precursor frequencies in peripheral blood lymphocytes. An increase in LAK precursor frequency above baseline was suggested by day 6 of this protocol and was clearly significant by day 20, indicating an expansion of the circulating precursor pool results from in vivo IL-2 administration. Correlations were not significant between LAK precursor frequency during IL-2 therapy and the total number of circulating lymphocytes, the percentage of CD56+ lymphocytes, IL-2 proliferative responses, or LAK activity of peripheral blood lymphocytes, indicating that the precursor frequency identification based on functional testing of individual cells is not accurately reflected by these analyses of heterogeneous bulk populations. Selective cell depletion analyses revealed that the majority of LAK precursors after in vivo IL-2 therapy were cells with the natural killer phenotype. Analysis of LAK precursors may help define the in vivo IL-2 administration, alone or in combination with other hematopoietic or immunodifferentiative cytokines, necessary to further augment in vivo effector cell numbers and activity for patients with cancer.


Asunto(s)
Interleucina-2/uso terapéutico , Células Asesinas Activadas por Linfocinas/efectos de los fármacos , Linfocitos/efectos de los fármacos , Neoplasias/sangre , Neoplasias/tratamiento farmacológico , Células Madre/efectos de los fármacos , División Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Evaluación de Medicamentos , Humanos , Recuento de Leucocitos/métodos , Subgrupos Linfocitarios/efectos de los fármacos
19.
Cancer Immunol Immunother ; 32(5): 325-30, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-1825620

RESUMEN

A total of 199 T cell clones from two melanoma patients were derived from progenitor T cells from recurrent melanoma, regional lymph nodes (either involved or uninvolved with malignancy) and peripheral blood by inoculating single cells directly into the wells of microtiter plates before in vitro expansion. The surface marker phenotype of most clones was CD4+CD8-, although some were CD4-CD8+. Genomic DNA prepared from all clones was analyzed by Southern blot hybridization using T cell receptor (TCR) beta and gamma gene probes, seeking clones with identical TCR gene rearrangement patterns as direct evidence for in vivo progenitor T cell clonal amplification. Probing HindIII-digested DNA with TCR beta and TCR gamma probes revealed several clones with identical TCR gene rearrangement patterns. These clones had subsequent probing of BamHI-digested DNA with TCR beta and TCR gamma probes, which showed all but 2 clones to have distinct rearrangement patterns. These analyses provide clear molecular evidence for in vivo polyclonal CD4+ T cell populations in each of several separate immune compartments in these patients.


Asunto(s)
Linfocitos Infiltrantes de Tumor/fisiología , Melanoma/sangre , Receptores de Antígenos de Linfocitos T/genética , Adulto , Southern Blotting , Antígenos CD4/inmunología , Células Cultivadas , Sondas de ADN/genética , Humanos , Ganglios Linfáticos/fisiología , Ganglios Linfáticos/ultraestructura , Activación de Linfocitos/fisiología , Subgrupos Linfocitarios/fisiología , Linfocitos/fisiología , Linfocitos/ultraestructura , Linfocitos Infiltrantes de Tumor/ultraestructura , Masculino , Melanoma/genética , Persona de Mediana Edad , Fenotipo , Receptores de Antígenos de Linfocitos T alfa-beta , Receptores de Antígenos de Linfocitos T gamma-delta , Linfocitos T/fisiología , Linfocitos T/ultraestructura
20.
Cancer Invest ; 9(1): 35-48, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-2012995

RESUMEN

Preliminary studies involving small numbers of patients have suggested that interleukin-2 (IL-2) administered by continuous infusion in repetitive weekly cycles using doses of 3 x 10(6) U/M2/day is immunologically active and can induce tumor responses in patients with renal cell carcinoma. This study was designed to examine both the immunological and clinical effects of prolonged infusion IL-2 given by repetitive weekly cycles; first at moderate doses for 4 weeks as an impatient followed by lower doses of IL-2 for up to 5 months. Prolonged IL-2 treatment was investigated because previous studies revealed that patients had a return to their baseline immune status within 4 weeks after completing IL-2 treatment. Twenty-five patients (including 18 with renal cell carcinoma) were treated with one of two regimens utilizing IL-2 as sole therapy. These regimens were designed to induce augmented and prolonged immune activation based upon in vitro and in vivo data. Though patients on both arms of the study demonstrated sustained lymphocytosis, increase in numbers of natural killer cells, and induction of lymphokine-activated killer activity with prolonged IL-2 administration, only 1 out of the 18 patients with renal cell carcinoma demonstrated a sustained partial antitumor response to therapy. Furthermore, several patients demonstrated profound immune activation, without any evidence of tumor regression. The lack of clinical responses in these patients showing marked activation of LAK cytotoxicity suggests that other variables must also influence the likelihood of antitumor effects for patients receiving IL-2 therapy.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Interleucina-2/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Adulto , Anciano , Carcinoma de Células Renales/inmunología , Citotoxicidad Inmunológica , Esquema de Medicación , Femenino , Humanos , Hipotensión/etiología , Interleucina-2/administración & dosificación , Interleucina-2/toxicidad , Neoplasias Renales/inmunología , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Naturales/inmunología , Leucocitosis/etiología , Masculino , Persona de Mediana Edad , Proyectos Piloto , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA