Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Appl Toxicol ; 36(2): 238-56, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26032931

RESUMEN

Combination of tumor antigens with immunostimulants is a promising approach in cancer immunotherapy. We assessed animal model toxicity of AS15 combined with various tumor antigens: WT1 (rabbits), or p501, dHER2 and recPRAME (cynomolgus monkeys), administered in seven or 20 dose regimens versus a saline control. Clinical and ophthalmological examinations, followed by extensive post-mortem pathological examinations, were performed on all animals. Blood hematology and biochemistry parameters were also assessed. Antigen-specific antibody titers were determined by enzyme-linked immunosorbent assay. Additional assessments in monkeys included electrocardiography and immunohistochemical evaluations of the p501 expression pattern. Transient increases in body temperature were observed 4 h or 24 h after injections of recPRAME + AS15 and dHER2 + AS15. Edema and erythema were observed up to 1 week after most injections of recPRAME + AS15 and all injections of dHER2 + AS15. No treatment-related effects were observed for electrocardiography parameters. Mean fibrinogen levels were significantly higher in all treated groups compared to controls, but no differences could be observed at the end of the treatment-free period. Transient but significant differences in biochemistry parameters were observed post-injection: lower albumin/globulin ratios (p501 + AS15), and higher bilirubin, urea and creatinine (dHER2 + AS15). Pathology examinations revealed significant increases in axillary lymph node mean weights (recPRAME + AS15) compared to controls. A 100% seroconversion rate was observed in all treated groups, but not in controls. p501 protein expression was observed in prostates of all monkeys from studies assessing p501 + AS15. These results suggest a favorable safety profile of the AS15-containing candidate vaccines, supporting the use of AS15 for clinical development of potential anticancer vaccines.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/uso terapéutico , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Adyuvantes Inmunológicos/toxicidad , Animales , Antígenos de Neoplasias/toxicidad , Femenino , Haplorrinos , Inyecciones Intramusculares , Masculino , Modelos Animales , Conejos
2.
Biochemistry ; 50(25): 5660-7, 2011 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-21595477

RESUMEN

Etoposide is a topoisomerase II poison that is used to treat a variety of human cancers. Unfortunately, 2-3% of patients treated with etoposide develop treatment-related leukemias characterized by 11q23 chromosomal rearrangements. The molecular basis for etoposide-induced leukemogenesis is not understood but is associated with enzyme-mediated DNA cleavage. Etoposide is metabolized by CYP3A4 to etoposide catechol, which can be further oxidized to etoposide quinone. A CYP3A4 variant is associated with a lower risk of etoposide-related leukemias, suggesting that etoposide metabolites may be involved in leukemogenesis. Although etoposide acts at the enzyme-DNA interface, several quinones poison topoisomerase II via redox-dependent protein adduction. The effects of etoposide quinone on topoisomerase IIα-mediated DNA cleavage have been examined previously. Although findings suggest that the activity of the quinone is slightly greater than that of etoposide, these studies were carried out in the presence of significant levels of reducing agents (which should reduce etoposide quinone to the catechol). Therefore, we examined the ability of etoposide quinone to poison human topoisomerase IIα in the absence of reducing agents. Under these conditions, etoposide quinone was ∼5-fold more active than etoposide at inducing enzyme-mediated DNA cleavage. Consistent with other redox-dependent poisons, etoposide quinone inactivated topoisomerase IIα when incubated with the protein prior to DNA and lost activity in the presence of dithiothreitol. Unlike etoposide, the quinone metabolite did not require ATP for maximal activity and induced a high ratio of double-stranded DNA breaks. Our results support the hypothesis that etoposide quinone contributes to etoposide-related leukemogenesis.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Benzoquinonas/metabolismo , Benzoquinonas/toxicidad , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Etopósido/metabolismo , Etopósido/toxicidad , Antígenos de Neoplasias/toxicidad , Catecoles/metabolismo , Aductos de ADN/efectos de los fármacos , Aductos de ADN/toxicidad , Daño del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/toxicidad , Proteínas de Unión al ADN/toxicidad , Ditiotreitol/toxicidad , Estabilidad de Enzimas/efectos de los fármacos , Etopósido/química , Humanos , Leucemia Mieloide Aguda/inducido químicamente , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Oxidación-Reducción , Sustancias Reductoras/farmacología
3.
Anticancer Res ; 39(4): 1699-1703, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30952708

RESUMEN

BACKGROUND/AIM: Previous work in rodent models showed that an autologous tissue vaccine is both a safe and effective approach for treating cancer; however, as a translational step, safety must first be evaluated in a more clinically-relevant model. MATERIALS AND METHODS: An autologous immunotherapy produced from resected tumors, was evaluated in a clinically-relevant canine model to assess safety. Ninety-three dogs with spontaneously occurring tumors received vaccination with inactivated autologous tumor tissue combined with an adjuvant of particulate porcine small intestinal submucosa extracellular matrix (SIS-ECM). Patients were followed to assess the occurrence of adverse events, overall survival, and tumor recurrence and/or metastasis. RESULTS: A small number (12%) of patients experienced limited, mild pyrexia, injection site swelling, or lethargy, all resolving without clinical intervention. CONCLUSION: Autologous whole cell cancer immunotherapy can be used safely in the canine model of cancer and represents a safe approach for the treatment for cancer.


Asunto(s)
Antígenos de Neoplasias/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Enfermedades de los Perros/terapia , Inmunoterapia/veterinaria , Neoplasias/veterinaria , Adyuvantes Inmunológicos , Animales , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/toxicidad , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/toxicidad , Enfermedades de los Perros/diagnóstico , Enfermedades de los Perros/inmunología , Enfermedades de los Perros/cirugía , Perros , Femenino , Inmunoterapia/efectos adversos , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Masculino , Márgenes de Escisión , Neoplasia Residual , Neoplasias/diagnóstico , Neoplasias/inmunología , Neoplasias/terapia , Sus scrofa
4.
Immunopharmacol Immunotoxicol ; 30(4): 747-60, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18720167

RESUMEN

We showed in a previous study that hot-water extracts of Agaricus blazei (Agaricus extracts) had anti-tumor activity to Meth A fibrosarcoma, but it remains unclear whether the Agaricus extracts ameliorate the skewed balance of type-1 T helper (Th1) and type-2 T helper (Th2) cells. We examined whether Agaricus extracts effect the skewed Th1/Th2 balance in tumor-bearing and asthma-induced mice. When Meth A-bearing mice were given orally either Agaricus extracts or water once a day starting 5 days after tumor implantation, spleen T cells, prepared from tumor-bearing mice treated with Agaricus extracts, in response to anti-CD3 monoclonal antibody produced significantly higher levels of interferon gamma (IFN-gamma) than that of controls. The mRNA expression of IFN-gamma-inducing protein 10 and the frequency of CD69(+) or CD49d(+) cells, among activated T cells infiltrated into tumors, significantly increased in Agaricus-treated mice, compared with those of tumor-controls. In asthma-induced mice, treatment with the Agaricus extracts caused significant downregulation of OVA-specific antibody responses of IgG1 and IgE but not of IgG2a, and significantly decreased total cell numbers, levels of interleukin 5, and eosinophil numbers in bronchial alveolar lavage fluids. IFN-gamma production by anti-CD3-stimulated spleen cells, obtained from Agaricus-treated mice, significantly increased. Our results strongly suggest that oral administration of Agaricus extracts ameliorates the Th1/Th2 balance from the Th2-skewed conditions.


Asunto(s)
Agaricus/inmunología , Asma/inmunología , Asma/terapia , Fibrosarcoma/inmunología , Fibrosarcoma/terapia , Células TH1/inmunología , Células Th2/inmunología , Administración Oral , Animales , Antiasmáticos/administración & dosificación , Antígenos de Neoplasias/toxicidad , Antineoplásicos/administración & dosificación , Asma/microbiología , Femenino , Fibrosarcoma/microbiología , Antígenos de Histocompatibilidad/toxicidad , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C , Células TH1/microbiología , Células TH1/patología , Células Th2/microbiología , Células Th2/patología
5.
Clin Cancer Res ; 10(16): 5391-402, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15328177

RESUMEN

PURPOSE: The tumor-associated antigen Ep-CAM (epithelial cell adhesion molecule) is overexpressed in colorectal carcinoma (CRC). The aim of the present study was to evaluate and compare the safety and immunogenicity of a recombinant Ep-CAM protein and a human anti-idiotypic antibody (anti-Id) mimicking Ep-CAM. EXPERIMENTAL DESIGN: Patients with resected American Joint Committee on Cancer stages II-IV CRC without remaining macroscopic disease received intradermal/subcutaneous injections of Ep-CAM (400 microg/dose; n = 7) or anti-Id (500 microg/dose; n = 6) at weeks 0, 2, and 6 in combination with granulocyte macrophage colony-stimulating factor (75 microg/day, for 4 consecutive days). RESULTS: Adverse reactions were mild (grade I-II). All patients immunized with the Ep-CAM protein produced Ep-CAM-specific IgG antibodies, predominantly IgG1 and IgG3 subclasses, whereas no humoral response was induced by the anti-Id vaccine. All patients, with one exception in each group, mounted an Ep-CAM-specific proliferative T-cell response. The immune response was more rapid, potent, and protracted after Ep-CAM in comparison with anti-Id vaccination. Interferon-gamma-secreting cells (ELISPOT) were detected in both immunization groups against the Ep-CAM protein as well as various Ep-CAM-derived MHC class I- and II-restricted peptides. Flow cytometry analysis showed that Ep-CAM-specific interferon-gamma- and perforin-producing cells predominantly resided within CD8(+)CD56- and CD8(dim)CD56+ T cells. CONCLUSIONS: Ep-CAM protein in combination with granulocyte macrophage colony-stimulating factor induced a long-lasting, Th1-biased humoral and cellular immune response compared with anti-Id. Ep-CAM-specific T cells and natural killer-like T cells responding in a MHC class I- and II-restricted manner were also induced. Vaccination with Ep-CAM protein may warrant further investigation as a novel therapeutic approach to CRC.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer , Moléculas de Adhesión Celular/inmunología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Anciano , Secuencia de Aminoácidos , Antígenos de Neoplasias/toxicidad , Moléculas de Adhesión Celular/toxicidad , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/terapia , Neoplasias Colorrectales/patología , Molécula de Adhesión Celular Epitelial , Epítopos/inmunología , Femenino , Humanos , Inmunoglobulina G/sangre , Masculino , Datos de Secuencia Molecular , Estadificación de Neoplasias , Fragmentos de Péptidos/química , Fragmentos de Péptidos/inmunología , Neoplasias del Recto/inmunología , Neoplasias del Recto/patología , Neoplasias del Recto/terapia
6.
J Immunother ; 38(8): 311-20, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26325375

RESUMEN

The PRAME tumor antigen is a potential target for immunotherapy. We assessed the immunogenicity, the antitumor activity, and the safety and the tolerability of a recombinant PRAME protein (recPRAME) combined with the AS15 immunostimulant (recPRAME+ AS15) in preclinical studies in mice and Cynomolgus monkeys. Four groups of 12 CB6F1 mice received 4 injections of phosphate-buffered saline (PBS), recPRAME, AS15, or recPRAME+AS15. Immunized mice were injected with tumor cells expressing PRAME (CT26-PRAME) 2 weeks or 2 months after the last injection. The mean tumor surface was measured twice a week. Two groups of 10 monkeys received 7 injections of saline or recPRAME+ AS15. T-cell responses were measured by flow cytometry using intracellular cytokine staining (ICS). In CB6F1 mice, repeated injections of recPRAME+ AS15 induced high PRAME-specific antibody titers and mostly CD4+ T cells producing cytokines. This immune response was long-lasting in these animals and was associated with protection against a challenge with PRAME-expressing tumor cells (CT26-PRAME) applied either 2 weeks or 2 months after the last injection; these data indicate the induction of an immune memory. In HLA-A02.01/HLA-DR1 transgenic mice, recPRAME+ AS15 induced both CD4+ and CD8+ T-cell responses, indicating that this antigen can be processed by the human leukocyte antigen and is potentially immunogenic in humans. In addition, a repeated-dose toxicity study in monkeys showed that 7 biweekly injections of recPRAME+ AS15 were well tolerated, and induced PRAME-specific antibodies and T cells. In conclusion, these preclinical data indicate that repeated injections of the PRAME cancer immunotherapeutic are immunogenic and have an acceptable safety profile.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Antígenos de Neoplasias/uso terapéutico , Neoplasias/tratamiento farmacológico , Adyuvantes Inmunológicos/toxicidad , Animales , Anticuerpos/sangre , Antígenos de Neoplasias/toxicidad , Línea Celular Tumoral , Quimioterapia Combinada , Femenino , Inmunoterapia , Macaca fascicularis , Masculino , Ratones , Ratones Transgénicos , Neoplasias/inmunología , Proteínas Recombinantes/uso terapéutico , Proteínas Recombinantes/toxicidad , Linfocitos T/inmunología
7.
Mutat Res ; 781: 37-48, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26421495

RESUMEN

Tyrosyl-DNA-phosphodiesterase 1 (TDP1) is a DNA repair enzyme that removes irreversible protein-linked 3' DNA complexes, 3' phosphoglycolates, alkylation damage-induced DNA breaks, and 3' deoxyribose nucleosides. In addition to its extended spectrum of substrates, TDP1 interacts with several DNA damage response factors. To determine whether TDP1 participates in the repair of topoisomerase II (Top2) induced DNA lesions, we generated TDP1 depleted (TDP1kd) human tumoral cells. We found that TDP1kd cells are hypersensitive to etoposide (ETO). Moreover, we established in a chromatin context that following treatment with ETO, TDP1kd cells accumulate increased amounts of Top2α cleavage complexes, removing them with an altered kinetics. We also showed that TDP1 depleted cells accumulate increased γH2AX and pS296Chk1 signals following treatment with ETO. Similarly, cytogenetics analyses following Top2 poisoning revealed increased amounts of chromatid and chromosome breaks and exchanges on TDP1kd cells in the presence or not of the DNA-PKcs inhibitor NU7026. However, the levels of sister chromatid exchanges were similar in both TDP1kd and control non-silenced cell lines. This suggests a role of TDP1 in both canonical non-homologous end joining and alternative end joining, but not in the homologous recombination repair pathway. Finally, micronucleus analyses following ETO treatment revealed a higher frequency of micronucleus containing γH2AX signals on TDP1kd cells. Together, our results highlight an active role of TDP1 in the repair of Top2-induced DNA damage and its relevance on the genome stability maintenance in human cells.


Asunto(s)
Antígenos de Neoplasias/toxicidad , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , ADN-Topoisomerasas de Tipo II/toxicidad , Proteínas de Unión al ADN/toxicidad , Hidrolasas Diéster Fosfóricas/metabolismo , Cromonas , Ensayo de Unidades Formadoras de Colonias , Reparación del ADN por Unión de Extremidades/fisiología , Cartilla de ADN/genética , Etopósido/farmacología , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Violeta de Genciana , Células HeLa , Histonas/metabolismo , Humanos , Immunoblotting , Pruebas de Micronúcleos , Morfolinas , Hidrolasas Diéster Fosfóricas/deficiencia , Hidrolasas Diéster Fosfóricas/genética , Proteínas de Unión a Poli-ADP-Ribosa , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Anticancer Res ; 18(4A): 2513-8, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9703902

RESUMEN

BACKGROUND: The murine monoclonal antibody NCC-ST-421 (ST-421) recognizes dimeric Le(a) antigen expressed on gastrointestinal cancer cells. MATERIALS AND METHODS: Direct antitumour activity of ST-421 was evaluated using dimeric Le(a) positive cell lines Colo 205, Colo 201, HT-29 and WiDr; and negative cell lines MX-1 and K562. RESULTS: While time- and concentration-dependent antitumour activity was observed against Colo 205 and Colo 201, no antitumour activity was detected against the other cell lines tested in an in vitro cytotoxicity assay. When ST-421 was administered intraperitoneally daily for 2 weeks to severe combined immunodeficient (SCID) mice transplanted with tumour xenografts, inhibition of tumour growth was observed against Colo 205, and to a lesser extent HT-29 and WiDr. Anti-asialo GM1 antibody did not block this antitumour activity, suggesting ST-421 has a direct cytotoxic effect. The degree of antitumour activity of ST-421 dependent on the grade of Le(a)-expression as detected by immunohistochemical staining. CONCLUSIONS: Flow cytometric and immunohistochemical analysis suggests the induction of apoptosis may play a key role in the direct antitumour activity of ST-421 on Colo 205 cells.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/uso terapéutico , Antineoplásicos/uso terapéutico , Apoptosis , Neoplasias del Colon/terapia , Animales , Anticuerpos Monoclonales/toxicidad , Antígenos de Neoplasias/toxicidad , Antineoplásicos/toxicidad , Biomarcadores de Tumor , Neoplasias de la Mama , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/patología , Femenino , Citometría de Flujo , Gangliósidos/inmunología , Humanos , Leucemia Eritroblástica Aguda , Masculino , Ratones , Ratones SCID , Trasplante Heterólogo , Células Tumorales Cultivadas
9.
Cancer Immunol Immunother ; 40(4): 219-27, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7538449

RESUMEN

In order to investigate tumoricidal effector cells in therapy by biological response modifiers (BRM) such as Propionibacterium acnes, bacillus Calmette-Guérin (BCG), Streptococcus pyogenes and a protein-bound polysaccharide (PSK), we established T cell lines specific for each BRM from BALB/c mice immunized with the corresponding BRM. These T cell lines proliferated and produced interleukin-2 (IL-2) and/or IL-4, but only in the presence of the relevant BRM and BALB/c spleen cells as the antigen and antigen-presenting cells respectively. Cross-functional experiments indicated that each BRM acts as a nominal antigen, but not as a non-specific immunostimulator. In addition, the T cell lines killed Ia-positive syngeneic B lymphoma cells, but only in the presence of the relevant BRM. These experiments excluded the possibility of cytotoxic effects by each BRM. The T cell lines and clones also killed Ia-negative bystander target cells, but only in the presence of both a relevant antigen and antigen-presenting cells. The T cell clones specific for S. pyogenes or P. acnes tested were Thy1+, L3T4+ and Lyt2-. These results indicate that some BRM exert tumoricidal activity by inducing T cells that recognize them as an antigen and kill tumor cells in an antigen-specific manner. The T cells killed tumor targets in either a tumor-necrosis-factor(TNF)-dependent or a TNF-independent manner. The mediator of the latter pathway remains to be elucidated.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/toxicidad , Antígenos/inmunología , Factores Inmunológicos/inmunología , Factores Inmunológicos/toxicidad , Inmunoterapia Adoptiva , Linfoma de Células B/terapia , Linfocitos T/inmunología , Animales , Vacuna BCG/inmunología , Vacuna BCG/toxicidad , Línea Celular , Epítopos , Inmunización , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Linfoma de Células B/inmunología , Ratones , Ratones Endogámicos BALB C , Propionibacterium acnes/inmunología , Proteoglicanos/inmunología , Proteoglicanos/toxicidad , Sensibilidad y Especificidad
10.
Mol Carcinog ; 3(4): 202-9, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2119594

RESUMEN

BALB/c 3T3 cells were exposed to 7,12-dimethylbenz[a]anthracene (DMBA) and resultant transformed foci were analyzed for the presence of A182----T mutation at codon 61 of Ha-ras (a mutation found in many DMBA-induced animal tumors). None of the 30 independently cloned transformed cell lines contained such a mutation. In order to see whether DMBA is able to induce this mutation in BALB/c 3T3 cells, we developed a method sensitive enough to detect this specific mutation at the frequency of 10(-6). Employing this assay, we found time- and dose-dependent induction by DMBA of Ha-ras A182----T mutation in BALB/c 3T3 cells; for example, 2 wk after exposure to 100 micrograms/mL DMBA, 1.4 in 1 X 10(4) cells contained this specific mutation. On the other hand, other agents that also induce BALB/c 3T3 cell transformation, such as 3-methylcholanthrene (MCA), 12-O-tetradecanoylphorbol-13-acetate (TPA), N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), or ultraviolet light, did not induce the mutation at detectable frequency (less than 10(-6)). These results suggest that DMBA efficiently induces Ha-ras mutation in BALB/c 3T3 cells but that this mutation is not recruited in the process of cell transformation. A hypothesis of carcinogen-specific mutation of Ha-ras gene and its tissue (cell type)-specific recruitment in carcinogenesis is proposed.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores , Carcinógenos , ADN/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genes ras , Mutación , Transformación Genética , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Antígenos de Neoplasias/toxicidad , Secuencia de Bases , Codón , Frecuencia de los Genes , Metilnitronitrosoguanidina/toxicidad , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Células Tumorales Cultivadas
11.
J Pathol ; 199(3): 398-408, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12579542

RESUMEN

The pancreatitis-associated protein (PAP) is a pancreatic stress protein overexpressed during acute pancreatitis, a disease often accompanied by lung inflammation. We investigated whether PAP was involved in the occurrence of this remote complication of pancreatitis and whether the liver might be implicated in the process. PAP was injected into the vena cava of rats (40 or 400 micro g/kg body weight). For comparison, pancreatitis was induced in rats by intraductal administration of sodium taurocholate. Three hours later, parameters of inflammation and mRNA concentrations of TNFalpha, P-selectin, heat shock protein (HSP)-70, and extracellular superoxide dismutase (EC-SOD) were monitored in lung and liver. Significant increases in P-selectin expression, neutrophil infiltration, and oxidative stress revealed that PAP treatment induced lung inflammation in rats and exacerbated inflammation in animals with pancreatitis. Plasma TNFalpha level was increased and TNFalpha mRNA was strongly overexpressed in liver, with concomitant activation of NF-kappaB; in situ hybridization revealed that TNFalpha overexpression was mainly located to hepatocytes. Lung inflammation induced by PAP could be prevented by injection of anti-TNFalpha antibodies. It was concluded that, during pancreatitis, PAP released by the pancreas could mediate lung inflammation through induction of hepatic TNFalpha expression and subsequent increase in circulating TNFalpha.


Asunto(s)
Antígenos de Neoplasias/toxicidad , Biomarcadores de Tumor/toxicidad , Hepatocitos/metabolismo , Neumonía/inducido químicamente , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Regulación de la Expresión Génica/efectos de los fármacos , Lectinas Tipo C , Masculino , FN-kappa B/metabolismo , Infiltración Neutrófila/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Selectina-P/metabolismo , Pancreatitis/inducido químicamente , Proteínas Asociadas a Pancreatitis , Neumonía/metabolismo , Neumonía/prevención & control , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética
12.
J Immunol ; 169(6): 3053-60, 2002 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-12218121

RESUMEN

We immunized mice with antigenic peptide P815E, which is presented by H-2K(d) and recognized by tumor-specific CTL raised against P815 tumor cells. This peptide is encoded by the ubiquitously expressed gene MsrA and carries a mutated residue conferring tumor specificity. Unexpectedly, we observed a severe toxicity occurring in the early hours after the third injection, resulting in the death of most mice within 24 h. The toxic syndrome was reminiscent of TNF-induced shock, and the sera of ill mice contained high levels of TNF. Toxicity was prevented by injection of neutralizing anti-TNF Abs, confirming the involvement of TNF. Depletion of CD8+ T cells could also prevent toxicity, and ex vivo experiments confirmed that CD8+ lymphocytes were the major cellular source of TNF in immunized mice. Tetramer analysis of the lymphocytes of immunized mice indicated a massive expansion of P815E-specific T cells, up to >60% of circulating CD8+ lymphocytes. A similar toxicity was observed after massive expansion of specific CD8+ T cells following immunization with another P815 peptide, which is encoded by gene P1A and was injected in a form covalently linked to an immunostimulatory peptide derived from IL-1. We conclude that the toxicity is caused by specific CD8+ lymphocytes, which are extensively amplified by peptide immunization in a QS21-based adjuvant and produce toxic levels of TNF upon further stimulation with the peptide. Our results suggest that immunotherapy trials involving new peptides should be pursued with caution and should include a careful monitoring of the T cell response.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Activación de Linfocitos , Péptidos/inmunología , Factor de Necrosis Tumoral alfa/fisiología , Factor de Necrosis Tumoral alfa/toxicidad , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/toxicidad , Linfocitos T CD8-positivos/metabolismo , Citotoxicidad Inmunológica/efectos de los fármacos , Epítopos de Linfocito T/inmunología , Inyecciones Subcutáneas , Activación de Linfocitos/efectos de los fármacos , Sarcoma de Mastocitos/inmunología , Ratones , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Péptidos/administración & dosificación , Péptidos/toxicidad , Choque Séptico/inmunología , Choque Séptico/mortalidad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
13.
J Immunol ; 169(9): 5227-35, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12391241

RESUMEN

TCR Id protein conjugated to keyhole limpet hemocyanin (KLH) (TCR Id:KLH) and injected with a chemical adjuvant (QS-21) induces a protective, Id-specific immune response against the murine T cell lymphoma, C6VL. However, Id-based immunotherapy of C6VL has not demonstrated therapeutic efficacy in tumor-bearing mice. We report here that C6VL lysate-pulsed dendritic cells (C6VL-DC) vaccines display enhanced efficacy in both the prevention and the therapy of T cell lymphoma compared with TCR Id:KLH with QS-21 vaccines. C6VL-DC vaccines stimulated potent tumor-specific immunity that protected mice against lethal challenge with C6VL and significantly enhanced the survival of tumor-bearing mice. Tumor-specific proliferation and secretion of IFN-gamma indicative of a Th1-type immune response were observed upon ex vivo stimulation of vaccine-primed lymph node cells. Adoptive transfer of immune T cell-enriched lymphocytes was sufficient to protect naive recipients from lethal tumor challenge. Furthermore, CD8(+) T cells were absolutely required for tumor protection. Although C6VL-DC and control vaccines stimulated low levels of tumor-specific Ab production in mice, Ab levels did not correlate with the protective ability of the vaccine. Thus, tumor cell lysate-pulsed DC vaccines appear to be an effective approach to generate potent T cell-mediated immune responses against T cell malignancies without requiring identification of tumor-specific Ags or patient-specific Id protein expression.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Fraccionamiento Celular , Células Dendríticas/trasplante , Idiotipos de Inmunoglobulinas/inmunología , Inmunoterapia Activa/métodos , Linfoma de Células T/inmunología , Linfoma de Células T/terapia , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/biosíntesis , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/toxicidad , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/toxicidad , Fraccionamiento Celular/métodos , Sistema Libre de Células/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Femenino , Esquemas de Inmunización , Idiotipos de Inmunoglobulinas/administración & dosificación , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Activación de Linfocitos/inmunología , Linfoma de Células T/mortalidad , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia , Linfocitos T/inmunología , Linfocitos T/trasplante , Células Tumorales Cultivadas
14.
Vaccine ; 19(27): 3652-60, 2001 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-11395199

RESUMEN

Human papillomavirus (HPV) E6 and E7 oncoproteins are attractive targets for T-cell-based immunotherapy of cervical intraepithelial neoplasia (CIN) and cancer. A newly designed vaccine, comprising the HPV16 L2, E6 and E7 as a single fusion protein (TA-CIN), was shown to elicit HPV16-specific CTL, T-helper cells and antibodies in a pre-clinical mouse model. These immune responses effectively prevented outgrowth of HPV16-positive tumour cells in a prophylactic setting as well as in a minimal residual disease setting. CTL immunity was optimally induced when TA-CIN was employed in heterologous prime-boost regimens in combination with TA-HPV, a clinical grade vaccinia-based vaccine. These data provide a scientific basis for the use of TA-CIN, alone or in combination with TA-HPV in future human trials.


Asunto(s)
Vacunas contra el Cáncer/toxicidad , Proteínas de la Cápside , Cápside/toxicidad , Proteínas Oncogénicas Virales/toxicidad , Papillomaviridae/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/uso terapéutico , Antígenos de Neoplasias/toxicidad , Antígenos Virales/administración & dosificación , Antígenos Virales/inmunología , Antígenos Virales/uso terapéutico , Antígenos Virales/toxicidad , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Cápside/administración & dosificación , Cápside/inmunología , Cápside/uso terapéutico , Línea Celular , Línea Celular Transformada , Evaluación Preclínica de Medicamentos , Inmunoterapia , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/uso terapéutico , Proteínas E7 de Papillomavirus , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/uso terapéutico , Vacunas Acelulares/administración & dosificación , Vacunas Acelulares/inmunología , Vacunas Acelulares/uso terapéutico , Vacunas Acelulares/toxicidad , Displasia del Cuello del Útero/prevención & control , Displasia del Cuello del Útero/terapia , Displasia del Cuello del Útero/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA