Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-32849281

RESUMEN

Medullary thyroid cancer (MTC) accounts for ~4% of all thyroid malignancies. MTC derives from the neural crest and secretes calcitonin (CTN) and carcinoembryonic antigen (CEA). Unlike differentiated thyroid cancer, MTC does not uptake iodine and I-131 RAI (radioactive iodine) treatment is ineffective. Patients with metastatic disease are candidates for FDA-approved agents with either vandetanib or cabozantinib; however, adverse effects limit their use. There are ongoing trials exploring the role of less toxic immunotherapies in patients with MTC. We present a 61-year-old male with the diagnosis of MTC and persistent local recurrence despite multiple surgeries. He was started on sunitinib, but ultimately its use was limited by toxicity. He then presented to the National Cancer Institute (NCI) and was enrolled on a clinical trial with heat-killed yeast-CEA vaccine (NCT01856920) and his calcitonin doubling time improved in 3 months. He then came off vaccine for elective surgery. After surgery, his calcitonin was rising and he enrolled on a phase I trial of avelumab, a programmed death-ligand 1 (PD-L1) inhibitor (NCT01772004). Thereafter, his calcitonin decreased > 40% on 5 consecutive evaluations. His tumor was subsequently found to express PD-L1. CEA-specific T cells were increased following vaccination, and a number of potential immune-enhancing changes were noted in the peripheral immunome over the course of sequential immunotherapy treatment. Although calcitonin declines do not always directly correlate with clinical responses, this response is noteworthy and highlights the potential for immunotherapy or sequential immunotherapy in metastatic or unresectable MTC.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígeno B7-H1/antagonistas & inhibidores , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/administración & dosificación , Carcinoma Neuroendocrino/tratamiento farmacológico , Saccharomyces cerevisiae/inmunología , Neoplasias de la Tiroides/tratamiento farmacológico , Vacunas de ADN/administración & dosificación , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma Neuroendocrino/inmunología , Carcinoma Neuroendocrino/patología , Quimioterapia Combinada , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Masculino , Persona de Mediana Edad , Pronóstico , Neoplasias de la Tiroides/inmunología , Neoplasias de la Tiroides/patología
2.
Int J Oncol ; 48(4): 1369-78, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26892612

RESUMEN

Vaccination with priming and expansion of tumour reacting T cells is an important therapeutic option to be used in combination with novel checkpoint inhibitors to increase the specificity of the T cell infiltrate and the efficacy of the treatment. In this phase I/II study, 14 high-risk disease-free ovarian (OC) and breast cancer (BC) patients after completion of standard therapies were vaccinated with MUC1, ErbB2 and carcinoembryonic antigen (CEA) HLA-A2+-restricted peptides and Montanide. Patients were subjected to 6 doses of vaccine every two weeks and a recall dose after 3 months. ECOG grade 2 toxicity was observed at the injection site. Eight out of 14 patients showed specific CD8+ T cells to at least one antigen. None of 4 patients vaccinated for compassionate use showed a CD8 activation. An OC patient who suffered from a lymph nodal recurrence, showed specific anti-ErbB2 CD8+ T cells in the bulky aortic lymph nodes suggesting homing of the activated T cells. Results confirm that peptide vaccination strategy is feasible, safe and well tolerated. In particular OC patients appear to show a higher response rate compared to BC patients. Vaccination generates a long-lasting immune response, which is strongly enhanced by recall administrations. The clinical outcome of patients enrolled in the trial appears favourable, having registered no deceased patients with a minimum follow-up of 8 years. These promising data, in line with the results of similar studies, the high compliance of patients observed and the favourable toxicity profile, support future trials of peptide vaccination in clinically disease-free patients who have completed standard treatments.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Antígeno Carcinoembrionario/administración & dosificación , Mucina-1/administración & dosificación , Neoplasias Ováricas/tratamiento farmacológico , Receptor ErbB-2/administración & dosificación , Adulto , Anciano , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Supervivencia sin Enfermedad , Femenino , Citometría de Flujo , Antígeno HLA-A2/genética , Antígeno HLA-A2/inmunología , Humanos , Inmunoterapia/métodos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Persona de Mediana Edad , Mucina-1/inmunología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Receptor ErbB-2/inmunología , Linfocitos T/inmunología
3.
Vaccine ; 29(47): 8642-8, 2011 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-21945963

RESUMEN

Protein vaccines may be a useful strategy for cancer immunotherapy because recombinant tumor antigen proteins can be produced on a large scale at relatively low cost and have been shown to be safe for clinical application. However, protein vaccines have historically exhibited poor immunogenicity; thus, an improved strategy is needed for successful induction of immune responses. TAT peptide is a protein transduction domain composed of an 11-amino acid peptide (TAT(47-57): YGRKKRRQRRR). The positive charge of this peptide allows protein antigen fused with it to improve cell penetration. Poly(I:C) is a synthetic double-stranded RNA that is negatively charged and favors interaction with the cationic TAT peptide. Poly(I:C) has been reported on adjuvant role in tumor vaccine through promotion of immune responses. Therefore, we demonstrated that vaccine with a mixture of TAT-CEA fusion protein and poly(I:C) can induce anti-tumor immunity in a murine colorectal tumor model. Splenocytes from mice vaccinated with a mixture of TAT-CEA fusion protein and poly(I:C) effectively induced CEA-specific IFN-γ-producing T cells and showed cytotoxic activity specific for MC-38-cea2 tumor cells expressing CEA. Vaccine with a mixture of TAT-CEA fusion protein and poly(I:C) delayed tumor growth in MC-38-cea-2 tumor-bearing mice. Depletion of CD8(+) T cells and NK cells reversed the inhibition of tumor growth in an MC-38-cea2-bearing mice, indicating that CD8(+) T cells and NK cells are responsible for anti-tumor immunity by vaccine with a mixture of TAT-CEA fusion protein and poly(I:C). Taken together, these results suggest that poly(I:C) could be used as a potent adjuvant to induce the anti-tumor immunity of a TAT-CEA fusion protein vaccine in a murine colorectal tumor model.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/prevención & control , Fragmentos de Péptidos/administración & dosificación , Poli I-C/administración & dosificación , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/administración & dosificación , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Femenino , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Procedimientos de Reducción del Leucocitos , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Bazo/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
4.
Vaccine ; 29(39): 6817-22, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21195077

RESUMEN

A plasmid DNA vaccine, encoding a truncated form of human CEA fused to a T-helper epitope (CEA66 DNA) was delivered three times intradermally at 2 mg or intramuscularly at 8 mg by Biojector® to patients with colorectal cancer. Prior to the first vaccination, all patients received cyclophosphamide (300 mg/m²) intravenously. Granulocyte-macrophage colony-stimulating factor (GM-CSF) was administered subcutaneously with each vaccination. All patients completed the vaccine schedule. There were no grade 3 or 4 adverse events (AE). The most frequently reported AE grades 1 and 2 were injection site reactions, fatigue, headache, arthralgia, chest tightness and myalgia. Vaccination with CEA66 DNA in combination with GM-CSF was well tolerated and no signs of autoimmunity have been detected.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/administración & dosificación , Neoplasias Colorrectales/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Vacunas de ADN/administración & dosificación , Anciano , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Neoplasias Colorrectales/inmunología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/inmunología , Sistemas de Liberación de Medicamentos/instrumentación , Evaluación de Medicamentos , Eritema/inducido químicamente , Femenino , Estudios de Seguimiento , Proteínas Ligadas a GPI/administración & dosificación , Proteínas Ligadas a GPI/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Humanos , Inyecciones Intradérmicas , Inyecciones Intramusculares , Masculino , Persona de Mediana Edad , Plásmidos/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Vacunación , Vacunas de ADN/efectos adversos , Vacunas de ADN/inmunología
5.
Int Immunopharmacol ; 9(6): 753-60, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19285575

RESUMEN

Involvement of the nitric oxide (NO) release in CEAM phi NLGP (carcinoembryonic antigen pulsed macrophages with neem leaf glycoprotein) vaccination and its relationship with vaccine induced type 1 immune response were aimed to study in the present communication. Vaccination with CEAM phi NLGP resulted in macrophage activation as evidenced by its increased number and expression of CD69 marker. Activated macrophages demonstrated upregulation in synthesis of IL-12 and downregulation in IL-10, along with excess IFN gamma production in splenic cells, as evidenced from mRNA analysis. Induction of such type 1 immunity was further confirmed by expression of type 1 specific transcription factor, T-bet and enhancement of intracellular glutathione content. Such vaccination also induced greater nitric oxide (NO) production from macrophages. Dependence of induced type 1 immune response on the NO release and vice versa was studied by in vitro neutralization of IFN gamma/IL-12 and in vivo inhibition of NO production by methylene blue. Obtained results clearly demonstrated the interdependence of two anti-tumor immune functions, namely, NO production and generation of type 1 immune response. Understanding of the mechanism of this NO related immune modulation would have great impact in proposing CEAM phi NLGP vaccine in clinic for the treatment of CEA+ tumors.


Asunto(s)
Adyuvantes Inmunológicos , Azadirachta/química , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Citocinas/biosíntesis , Glicoproteínas/administración & dosificación , Macrófagos Peritoneales/inmunología , Óxido Nítrico/biosíntesis , Animales , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/administración & dosificación , Femenino , Glutatión/inmunología , Glutatión/metabolismo , Glicoproteínas/química , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Activación de Macrófagos , Ratones , Hojas de la Planta/química , Hojas de la Planta/inmunología , Proteínas de Dominio T Box/inmunología , Proteínas de Dominio T Box/metabolismo , Vacunación
6.
BMC Cancer ; 9: 66, 2009 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-19243606

RESUMEN

BACKGROUND: Advanced pancreatic cancer has a poor prognosis, and the current standard of care (gemcitabine based chemotherapy) provides a small survival advantage. However the drawback is the accompanying systemic toxicity, which targeted treatments may overcome. This study aimed to evaluate the safety and tolerability of KAb201, an anti-carcinoembryonic antigen monoclonal antibody, labelled with I(131) in pancreatic cancer (ISRCTN 16857581). METHODS: Patients with histological/cytological proven inoperable adenocarcinoma of the head of pancreas were randomised to receive KAb 201 via either the intra-arterial or intravenous delivery route. The dose limiting toxicities within each group were determined. Patients were assessed for safety and efficacy and followed up until death. RESULTS: Between February 2003 and July 2005, 25 patients were enrolled. Nineteen patients were randomised, 9 to the intravenous and 10 to the intra-arterial arms. In the intra-arterial arm, dose limiting toxicity was seen in 2/6 (33%) patients at 50 mCi whereas in the intravenous arm, dose limiting toxicity was noted in 1/6 patients at 50 mCi, but did not occur at 75 mCi (0/3).The overall response rate was 6% (1/18). Median overall survival was 5.2 months (95% confidence interval = 3.3 to 9 months), with no significant difference between the intravenous and intra-arterial arms (log rank test p = 0.79). One patient was still alive at the time of this analysis. CONCLUSION: Dose limiting toxicity for KAb201 with I(131) by the intra-arterial route was 50 mCi, while dose limiting toxicity was not reached in the intravenous arm.


Asunto(s)
Adenocarcinoma/diagnóstico por imagen , Antígeno Carcinoembrionario/administración & dosificación , Inmunotoxinas/administración & dosificación , Radioisótopos de Yodo/administración & dosificación , Neoplasias Pancreáticas/radioterapia , Radioinmunoterapia/métodos , Adenocarcinoma/inmunología , Anciano , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/inmunología , Antígeno Carcinoembrionario/efectos adversos , Antígeno Carcinoembrionario/inmunología , Humanos , Inmunotoxinas/efectos adversos , Inmunotoxinas/inmunología , Inmunotoxinas/farmacocinética , Infusiones Intraarteriales , Infusiones Intravenosas , Radioisótopos de Yodo/efectos adversos , Radioisótopos de Yodo/farmacocinética , Persona de Mediana Edad , Neoplasias Pancreáticas/inmunología , Radiografía , Radioinmunoterapia/efectos adversos , Tasa de Supervivencia
7.
Cancer Sci ; 99(5): 1034-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18294279

RESUMEN

Although dendritic cells (DC) have been well demonstrated as a strong cellular adjuvant for a tumor vaccine, there are several limitations for clinical application. A protein-based vaccine using a potent adjuvant is an appealing approach for tumor antigen-specific immunotherapy because of their simplicity, safety, efficacy and capacity for repeated administration. CpG-oligodeoxynucleotides (ODN) have been used as adjuvants to stimulate innate and adaptive immune responses for cancer treatment. The authors evaluated the adjuvant effects of CpG-ODN in a vaccine incorporating recombinant fusion protein of the HIV TAT PTD domain and carcinoembryonic antigen (TAT-CEA). Mice vaccinated with TAT-CEA and CpG-ODN (TAT-CEA + CpG) showed enhanced CEA-specific immunity, including cytotoxic T-lymphocytes (CTL) activity and interferon (IFN)-gamma secreting T cells compared with CEA and CpG-ODN (CEA + CpG) or TAT-CEA vaccination alone. Vaccination with TAT-CEA + CpG elicited Th1-based responses, as indicated by the higher ratio of immunoglobulin (Ig)G2a antibody/IgG1 antibodies specific for CEA. The survival rate was significantly increased after vaccination with TAT-CEA + CpG in a tumor model using MC38/CEA2. Furthermore, the TAT-CEA +/- CpG vaccine groups showed similar antitumor immunity to the CEA peptide-pulsed DC (CEA peptide/DC) vaccine groups. These data suggest that coadministration of TAT fusion protein with CpG-ODN may serve as a potential formulation for enhancing antitumor activity.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Proteínas Reguladoras de la Apoptosis/uso terapéutico , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Oligodesoxirribonucleótidos/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/uso terapéutico , Adyuvantes Inmunológicos/administración & dosificación , Animales , Proteínas Reguladoras de la Apoptosis/administración & dosificación , Vacunas contra el Cáncer/uso terapéutico , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/uso terapéutico , Femenino , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Survivin , Linfocitos T Citotóxicos/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/administración & dosificación
8.
J Immunol ; 174(10): 5994-6004, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15879092

RESUMEN

Recent studies in both animal models and clinical trials have demonstrated that the avidity of T cells is a major determinant of antitumor and antiviral immunity. In this study, we evaluated several different vaccine strategies for their ability to enhance both the quantity and avidity of CTL responses. CD8(+) T cell quantity was measured by tetramer binding precursor frequency, and avidity was measured by both tetramer dissociation and quantitative cytolytic function. We have evaluated a peptide, a viral vector expressing the Ag transgene alone, with one costimulatory molecule (B7-1), and with three costimulatory molecules (B7-1, ICAM-1, and LFA-3), with anti-CTLA-4 mAb, with GM-CSF, and combinations of the above. We have evaluated these strategies in both a foreign Ag model using beta-galactosidase as immunogen, and in a "self" Ag model, using carcinoembryonic Ag as immunogen in carcinoembryonic Ag transgenic mice. The combined use of several of these strategies was shown to enhance not only the quantity, but, to a greater magnitude, the avidity of T cells generated; a combination strategy is also shown to enhance antitumor effects. The results reported in this study thus demonstrate multiple strategies that can be used in both antitumor and antiviral vaccine settings to generate higher avidity host T cell responses.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Pruebas Inmunológicas de Citotoxicidad/métodos , Activación de Linfocitos/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adyuvantes Inmunológicos/genética , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD , Antígenos de Diferenciación/inmunología , Antígeno CTLA-4 , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Terapia Combinada , Femenino , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Unión Proteica/inmunología , beta-Galactosidasa/administración & dosificación , beta-Galactosidasa/genética , beta-Galactosidasa/inmunología
9.
Vaccine ; 23(27): 3565-71, 2005 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-15855015

RESUMEN

We have studied the effects of recombinant adenoviruses as immune adjuvants for DNA vaccination. In a mouse model, using the weak immunogen carcinoembryonic antigen (CEA), anti-CEA IgG production was significantly higher and occurred earlier when immunization included a recombinant adenovirus together with CEA-plasmid DNA. Combined immunization with a recombinant adenovirus expressing the immunomodulatory molecule heat shock protein 72 (ADHSP72) and CEA-plasmid DNA resulted in CEA-specific T-cell activation capable of protecting mice from tumor formation with CEA expressing cells. Additionally, animals with CEA expressing tumors showed diminished tumor growth and prolonged survival when immunized with ADHSP72 and CEA-plasmid DNA compared to controls. Recombinant adenoviruses expressing immunomodulatory molecules such as HSP72 may be useful adjuvants for DNA vaccination.


Asunto(s)
Adenovirus Humanos/inmunología , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/inmunología , Proteínas de Choque Térmico/inmunología , Plásmidos , Neoplasias de la Próstata/inmunología , Adenovirus Humanos/genética , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/genética , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/genética , Femenino , Proteínas del Choque Térmico HSP72 , Proteínas de Choque Térmico/administración & dosificación , Proteínas de Choque Térmico/genética , Humanos , Inmunoglobulina G/biosíntesis , Memoria Inmunológica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante de Neoplasias , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/prevención & control , Análisis de Supervivencia , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
10.
J Immunol ; 174(5): 2645-52, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15728471

RESUMEN

Most tumor-associated Ags are self proteins that fail to elicit a T cell response as a consequence of immune tolerance. Dendritic cells (DCs) generated ex vivo have been used to break tolerance against such self Ags; however, in vitro manipulation of DCs is cumbersome and difficult to control, resulting in vaccines of variable potency. To address this problem we developed a method for loading and activating DCs, in situ, by first directing sufficient numbers of DCs to peripheral tissues using Flt3 ligand and then delivering a tumor-associated Ag and oligonucleotide containing unmethylated CG motifs to these tissues. In this study, we show in three different tumor models that this method can overcome tolerance and induce effective antitumor immunity. Vaccination resulted in the generation of CD8(+) T and NK cell effectors that mediated durable tumor responses without attacking normal tissues. These findings demonstrate that unmodified tumor-associated self Ags can be targeted to DCs in vivo to induce potent systemic antitumor immunity.


Asunto(s)
Antígenos de Neoplasias/inmunología , Autoantígenos/inmunología , Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Células Dendríticas/inmunología , Tolerancia Inmunológica , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Animales , Antígenos de Neoplasias/administración & dosificación , Autoantígenos/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Proliferación Celular , Neoplasias del Colon/patología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Células Asesinas Naturales/inmunología , Masculino , Melanoma Experimental/patología , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Oligodesoxirribonucleótidos/administración & dosificación , Oligodesoxirribonucleótidos/inmunología , Oxidorreductasas/administración & dosificación , Oxidorreductasas/inmunología , Proteínas Oncogénicas de Retroviridae/administración & dosificación , Proteínas Oncogénicas de Retroviridae/inmunología , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología
11.
Biochem Biophys Res Commun ; 328(1): 98-105, 2005 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-15670756

RESUMEN

Integration of lymphocyte-activating cytokines (e.g., interleukin-12: IL-12) to tumor cells offers promise for cancer immunotherapy, but the preparation of such heterodimeric proteins by refolding is difficult because of subunit instability. We achieved the refolding of Escherichia coli-expressed human IL-12 by a stepwise dialysis method, preventing the formation of insoluble aggregates by adding a redox reagent and an aggregation suppressor. We also constructed a tumor-specific IL-12 protein, each subunit of which was fused with one chain of variable domain fragment (Fv) of anticarcinoembryonic antigen (CEA) antibody T84.66 (aCEA-IL12). Fusion of IL-12 with Fv greatly increased the yield of functional heterodimer. Several assays have indicated that the Fv domain and IL-12 domain of the fused protein had cognate biological activities, and it enhanced the cytotoxicity of T-LAK cells for the cancer cell line.


Asunto(s)
Anticuerpos/administración & dosificación , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Antígeno Carcinoembrionario/administración & dosificación , Interleucina-12/administración & dosificación , Activación de Linfocitos/efectos de los fármacos , Ingeniería de Proteínas/métodos , Animales , Anticuerpos/genética , Anticuerpos/inmunología , Neoplasias de los Conductos Biliares/inmunología , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/administración & dosificación , Citocinas/química , Citocinas/genética , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Humanos , Interleucina-12/química , Interleucina-12/genética , Interleucina-12/inmunología , Microdiálisis/métodos , Pliegue de Proteína , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química
12.
Rev. bras. colo-proctol ; 23(3): 163-167, set. 2003. tab
Artículo en Portugués | LILACS | ID: lil-357983

RESUMEN

Os marcadores biológicos representam os exames de primeira linha na investigação das recidivas neoplásicas. No câncer colorretal, a relação custo-benefício da realização simultânea das dosagens séricas de CEA, fosfatase alcalina (FA) e gama-glutamil-transpeptidase (GT) e a validade de se continuar dosando CEA, quando o exame inicial é normal,não estão claras na literatura. Com o objetivo de estudar o comportamento desses marcadores na evolução dos tumores colorretais, foram estudados 100 pacientes,acompanhados no PO por tempo médio de 30 meses,no período de janeiro de 1991 a fevereiro de 1999 (8 anos). Cinqüenta e três eram do sexo masculino e quarenta e sete do feminino,com idade média de 60 anos. Todos os pacientes foram submetidos a tratamento cirúrgico,sendo que 12 já apresentavam metástases e 17 apresentaram recidiva durante o seguimento. Os resultados mostraram sensibilidade e especificidade de CEA, FA e GT em detectar metástases na operação de, respectivamente, 75por cento; 64por cento; 25por cento; 93por cento; 36por cento; 86por cento e em recidivas de 93por cento; 98por cento; 36por cento; 76por cento; 42por cento; 80por cento, respectivamente. O CEA quase sempre se elevou meses antes que FA e -GT, exceto em um paciente com metástases e noutro com recidiva. Dos pacientes que apresentaram metástases ou recidivas 45,9por cento tinham CEA inicial aumentado e somente 19,2por cento tinham CEA normal. Todos os pacientes que apresentaram CEA normal no Pré-OP,tiveram elevação desse marcador quando se diagnosticou a recidiva,exceto no paciente nº12. Concluiu-se que não há benefício na dosagem da FA e da dGT para diagnóstico de metástases hepáticas e que o CEA deve fazer parte do rastreamento de metástases, mesmo quando o seu valor inicial for normal.


Asunto(s)
Humanos , Anciano , Fosfatasa Alcalina , Antígeno Carcinoembrionario/administración & dosificación , Neoplasias Colorrectales , gamma-Glutamiltransferasa
13.
J Immunol ; 169(10): 5780-6, 2002 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-12421958

RESUMEN

A new strategy to improve the therapeutic utility of redirected T cells for cancer involves the development of novel Ag-specific chimeric receptors capable of stimulating optimal and sustained T cell antitumor activity in vivo. Given that T cells require both primary and costimulatory signals for optimal activation and that many tumors do not express critical costimulatory ligands, modified single-chain Ab receptors have been engineered to codeliver CD28 costimulation. In this study, we have compared the antitumor potency of primary T lymphocytes expressing carcinoembryonic Ag (CEA)-reactive chimeric receptors that incorporate either TCR-zeta or CD28/TCR-zeta signaling. Although both receptor-transduced T cell effector populations demonstrated cytolysis of CEA(+) tumors in vitro, T cells expressing the single-chain variable fragment of Ig (scFv)-CD28-zeta chimera had a far greater capacity to control the growth of CEA(+) xenogeneic and syngeneic colon carcinomas in vivo. The observed enhanced antitumor activity of T cells expressing the scFv-CD28-zeta receptor was critically dependent on perforin and the production of IFN-gamma. Overall, this study has illustrated the ability of a chimeric scFv receptor capable of harnessing the signaling machinery of both TCR-zeta and CD28 to augment T cell immunity against tumors that have lost expression of both MHC/peptide and costimulatory ligands in vivo.


Asunto(s)
Antígenos CD28/inmunología , Neoplasias del Colon/prevención & control , Rechazo de Injerto/inmunología , Región Variable de Inmunoglobulina/inmunología , Inmunoterapia Adoptiva/métodos , Receptores Inmunológicos/administración & dosificación , Receptores Inmunológicos/biosíntesis , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/biosíntesis , Adyuvantes Inmunológicos/genética , Animales , Sitios de Unión de Anticuerpos/genética , Antígenos CD28/administración & dosificación , Antígenos CD28/genética , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/inmunología , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Citotoxicidad Inmunológica/genética , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/metabolismo , Rechazo de Injerto/genética , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/biosíntesis , Inhibidores de Crecimiento/genética , Humanos , Región Variable de Inmunoglobulina/administración & dosificación , Región Variable de Inmunoglobulina/genética , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/fisiología , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Perforina , Proteínas Citotóxicas Formadoras de Poros , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Inmunológicos/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Linfocitos T Citotóxicos/trasplante
14.
Eur J Cancer ; 35(5): 815-24, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10505044

RESUMEN

The temporal variation in bone marrow proliferation has been used to help define the optimal time of day to dose with approximately 30 chemotherapeutic agents, so that treatment efficacy is maximised and toxicity is minimised. Since myelosuppression is also the dose-limiting toxicity for most forms of radioimmunotherapy, we hypothesised that time of day of administration might also influence tolerance for radioantibody therapy. Bone marrow proliferative activity in BALB/c mice was determined using cell cycle analysis of propidium iodide-stained bone marrow samples collected at 3 h intervals. Myelosuppression was determined at weekly intervals after a therapeutic dose of 131I-NP-4 anti-CEA (carcinoembryonic antigen) intact IgG at either 0900 h (2 h after light onset [HALO]), 1300 h (6 HALO) or 1600 h (9 HALO). The highest bone marrow proliferative activity was noted between 20 HALO (0300 h) and 4 HALO (1100 h), and the lowest activity could be measured at 10-13 HALO (1700-2000 h). Seven days after a maximal tolerated dose (MTD) of radioantibody, granulocyte reduction was 50% at both 2 and 6 HALO and only 32% at 9 HALO (P < 0.003). Fourteen days after radioantibody therapy, an 87% granulocyte suppression was observed in mice treated at 2 HALO and only a 64% granulocyte loss was noted in the 9 HALO treated group (P < 0.001). The 2 HALO group recovered earlier than the 9 HALO group (P < 0.013; 22% loss from the 2 HALO dose and 40% loss from the 9 HALO dose) on day 28 post-radioimmunotherapy. The difference in magnitude of neutropenia, rather than duration, was critical for establishing the MTD. A 30% increase in the MTD was possible if mice were dosed at 9 HALO (320 microCi) versus 2 HALO (240 microCi). These studies suggest that principles of chronobiology may govern the magnitude of toxicity and the highest dose tolerated in radioantibody therapy in the same way that it does for cytotoxic drug therapy.


Asunto(s)
Antígeno Carcinoembrionario/administración & dosificación , Inmunoglobulina G/administración & dosificación , Radioisótopos de Yodo/farmacocinética , Radioinmunoterapia/métodos , Animales , Células de la Médula Ósea/efectos de los fármacos , Antígeno Carcinoembrionario/inmunología , Ciclo Celular , Ritmo Circadiano , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos BALB C , Factores de Tiempo
15.
Clin Cancer Res ; 5(9): 2330-7, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10499601

RESUMEN

The principal objectives of this trial were twofold: (a) to examine the safety and relative efficacy of intradermal needle injection versus s.c. jet administration of a carcinoembryonic antigen (CEA)-encoding recombinant vaccinia virus (rV-CEA) over a limited dose range and (b) to evaluate CEA-specific immune responses or antitumor effects induced by rV-CEA vaccination. Patients were randomly assigned to one of two groups, depending upon the technique of vaccine administration. All 20 patients received two doses of 10(7) or 10(8) pfu of rV-CEA at a 4-week interval. Toxicity was limited to modest local inflammation at the inoculation site as well as low-grade fever and increased fatigue, each affecting fewer than 20% of the patients. No evidence of CEA-specific lymphoproliferation, interleukin 2 release, delayed-type hypersensitivity, or antibody response was observed. Thus, the efficacy comparison between the two administration techniques was based upon the induction of immune responses to the vaccinia virus vector. Both techniques induced vaccinia-specific lymphoproliferation, interleukin 2 release, and antibody responses of comparable magnitude and frequency as well as protected 80% of patients against pustule formation following vaccinia scarification. Thus, there is no compelling reason to recommend one administration technique over the other based upon toxicity or efficacy. We have selected s.c. jet injection for subsequent trials of rV-CEA based on the ability to accommodate larger injection volumes, enhanced standardization between clinicians, and avoidance of needles that could transmit the vaccine or blood-borne pathogens to health care workers. We recommend use of 10(8) pfu doses for subsequent trials of recombinant vaccinia virus vaccines based upon the favorable toxicity profile and more consistent local pustule formation indicative of an adequate inoculation of live virus. No objective clinical responses to the rV-CEA vaccine were observed among this population of patients with widely metastatic adenocarcinoma.


Asunto(s)
Adenocarcinoma/secundario , Adenocarcinoma/terapia , Vacunas contra el Cáncer/administración & dosificación , Antígeno Carcinoembrionario/genética , Virus Vaccinia/genética , Adenocarcinoma/inmunología , Adulto , Anciano , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/biosíntesis , Antígeno Carcinoembrionario/inmunología , Femenino , Humanos , Inyecciones Intradérmicas , Inyecciones Subcutáneas , Interleucina-2/inmunología , Interleucina-2/metabolismo , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Virus Vaccinia/inmunología
18.
Ann N Y Acad Sci ; 716: 104-14; discussion 140-3, 1994 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-8024189

RESUMEN

A gene therapy approach has been described that generates a tumor-selective qualitative difference in the metabolic capability in tumor cells. This is the result of the selective expression of a nonmammalian enzyme in tumor cells. Selective expression is achieved by utilization of a chimeric gene composed of the TRS from a tumor-associated marker gene linked to the coding domain of a gene encoding a nonmammalian enzyme. We have described the application of this approach for the treatment of metastatic CRC. This approach involves creation of a chimeric gene composed of the CEA TRS linked to the coding domain of the CD gene. Selective expression of CD in the tumor cells will allow the selective conversion of the prodrug 5-FCyt to 5-FCyt in the tumor while sparing normal cells. Most importantly, delivery and expression of CD into a small fraction of tumor cells may be sufficient to achieve a significant antitumor effect.


Asunto(s)
Neoplasias Colorrectales/terapia , Enzimas/administración & dosificación , Técnicas de Transferencia de Gen , Profármacos/metabolismo , Animales , Proteínas Bacterianas/administración & dosificación , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/genética , Carcinoma/terapia , Clonación Molecular , Citosina Desaminasa , Humanos , Neoplasias Hepáticas/secundario , Ratones , Nucleósido Desaminasas/administración & dosificación , Nucleósido Desaminasas/genética
19.
Rev. Inst. Nac. Enfermedades Respir ; 6(3): 128-34, jul.-sept. 1993. tab
Artículo en Español | LILACS | ID: lil-134870

RESUMEN

Tres de los más grandes problemas que afrontan el médico ante el cáncer broncogénico son, en primer lugar, su diagnóstico temprano, secundariamente el tratamiento específico y, finalmente, medios adecuados para determinar la evolutividad del padecimiento. Por ello, muy probablemente los marcadores tumorales, en un futuro ocupen un lugar preponderante para mejorar la sobrevida, calidad de la misam y el tratamiento adecuado en los pacientes portadores de neoplasias pulmonares. El presente trabajo, está avalado por la experiencia de nuestro Departamento con dos marcadores tumorales: antígeno carcinoembrionario y hormona gonadotropina criónica en 20 pacientes con otras patologías pulmonares. Nuestros resultados demostraron significación estadística, aunque no en todos los rubros estudiados, de lo que se concluye que es necesario un mayor número de investigaciones en cuanto a cáncer pulmonar y marcadores tumorales re refiere; tomando en cuenta que en la actualidad se conocen más de 20 marcadores diversos, sin que ninguno reúne un alto grado de confiabilidad


Asunto(s)
Humanos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Antígeno Carcinoembrionario , Carcinoma Broncogénico/diagnóstico , Gonadotropina Coriónica , Biomarcadores de Tumor , Antígeno Carcinoembrionario/administración & dosificación , Carcinoma Broncogénico/epidemiología , Carcinoma Broncogénico/terapia , Gonadotropina Coriónica/administración & dosificación
20.
Br J Cancer ; 67(3): 464-70, 1993 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8439497

RESUMEN

Carcinoembryonic antigen (CEA) is an oncofetal antigen whose function in the progression of colorectal carcinoma remains unclear although recent studies suggest it participates in homotypic cellular adhesion. We have previously shown that 40 micrograms of CEA injected intravenously into athymic nude mice enhances experimental metastasis in liver and lung by two human colorectal carcinoma cell lines that are injected intrasplenically 30 min later. The metastatic potential of another three moderately to highly metastatic colorectal carcinoma cell lines and of one weakly metastatic line has now been analysed in this model. CEA pretreatment only enhanced colony formation by cell lines that were weakly metastatic in untreated nude mice; it did not affect experimental metastasis by highly metastatic lines. CEA pretreatment enhanced the retention of 125I Idudr-labelled weakly metastatic tumour cells within the liver and lungs 4 h after intrasplenic injection but not the retention of highly metastatic tumour cells or inert latex beads. A significant correlation existed between the formation of experimental metastases and the early retention of tumour cells within the liver after intrasplenic injection. Aggregation did not appear to be important for retention in liver because CEA did not aggregate colorectal carcinoma cells in vitro. Also CEA did not alter natural host effector cell function in a cytolysis assay in vitro. We suggest that CEA facilitates liver colonisation by three of eight human colorectal carcinomas in athymic nude mice by increasing the hepatic retention of tumour cells. The potential mechanisms by which CEA may increase the retention of tumour cells in the liver are discussed.


Asunto(s)
Antígeno Carcinoembrionario/farmacología , Neoplasias del Colon , Neoplasias Hepáticas/secundario , Neoplasias del Recto , Animales , Antígeno Carcinoembrionario/administración & dosificación , Antígeno Carcinoembrionario/análisis , Femenino , Humanos , Inyecciones Intravenosas , Macrófagos del Hígado/química , Hígado/química , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Bazo/inmunología , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...