Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Protein Expr Purif ; 134: 72-81, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28389350

RESUMEN

The trematode Schistosoma mansoni Sm14 antigen was expressed in the yeast Pichia pastoris and secreted into the culture medium at yields of approximately 250 mg L-1. Sm14 belongs to a family of fatty-acid binding proteins and appears to play an important role in uptake, transport, and compartmentalization of lipids in S. mansoni and it is a potential vaccine candidate in both humans and domesticated animals. The Sm14 gene was codon-optimized for expression in P. pastoris, and placed under transcription of the strong methanol inducible AOX1 promoter. Mut+ transformants were selected and used in fed-batch cultivation using a 2.5L fermenter equipped with an on-line methanol control system in order to maintain constant methanol levels during induction. Optimal conditions for the expression of Sm14 by P. pastoris were found to be: dissolved oxygen at 40%, temperature of 25 °C, pH 5.0, and a constant methanol concentration of 1 gL-1. Our results show that a correctly processed Sm14 was secreted into the culture medium at levels of approximately 250  mg L-1. Sm14 from clarified culture medium was purified using a two-step procedure: anion-exchange chromatography followed by hydrophobic interaction chromatography, resulting in >95% purity with a final yield of 40% from the starting cell culture medium. This product has been tested in preliminary clinical trials and shown to elicit an antibody response with no adverse reactions.


Asunto(s)
Antígenos Helmínticos , Proteínas de Transporte de Ácidos Grasos , Expresión Génica , Proteínas del Helminto , Pichia/metabolismo , Schistosoma mansoni/genética , Vacunas , Animales , Antígenos Helmínticos/biosíntesis , Antígenos Helmínticos/genética , Antígenos Helmínticos/inmunología , Antígenos Helmínticos/aislamiento & purificación , Proteínas de Transporte de Ácidos Grasos/biosíntesis , Proteínas de Transporte de Ácidos Grasos/genética , Proteínas de Transporte de Ácidos Grasos/inmunología , Proteínas de Transporte de Ácidos Grasos/aislamiento & purificación , Proteínas del Helminto/biosíntesis , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Proteínas del Helminto/aislamiento & purificación , Humanos , Pichia/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Schistosoma mansoni/inmunología , Vacunas/biosíntesis , Vacunas/genética , Vacunas/inmunología , Vacunas/aislamiento & purificación
2.
Cancer Immunol Immunother ; 64(3): 311-23, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25403749

RESUMEN

The development of cancer immunotherapy has long been a challenge. Here, we report that prophylactic vaccination with a highly attenuated Trypanosoma cruzi strain expressing NY-ESO-1 (CL-14-NY-ESO-1) induces both effector memory and effector CD8(+) T lymphocytes that efficiently prevent tumor development. However, the therapeutic effect of such a vaccine is limited. We also demonstrate that blockade of Cytotoxic T Lymphocyte Antigen 4 (CTLA-4) during vaccination enhances the frequency of NY-ESO-1-specific effector CD8(+) T cells producing IFN-γ and promotes lymphocyte migration to the tumor infiltrate. As a result, therapy with CL-14-NY-ESO-1 together with anti-CTLA-4 is highly effective in controlling the development of an established melanoma.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Vacunas contra el Cáncer/inmunología , Inmunoterapia/métodos , Melanoma Experimental/terapia , Proteínas de la Membrana/inmunología , Animales , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/genética , Linfocitos T CD8-positivos/parasitología , Antígeno CTLA-4/antagonistas & inhibidores , Femenino , Humanos , Melanoma Experimental/inmunología , Melanoma Experimental/parasitología , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Trypanosoma cruzi/genética , Trypanosoma cruzi/inmunología
3.
Eur J Nucl Med Mol Imaging ; 42(11): 1700-1706, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26194713

RESUMEN

PURPOSE: The molecular specificity of monoclonal antibodies (mAbs) directed against tumor antigens has proven effective for targeted therapy of human cancers, as shown by a growing list of successful antibody-based drug products. We describe a novel, nonlinear compartmental model using PET-derived data to determine the "best-fit" parameters and model-derived quantities for optimizing biodistribution of intravenously injected (124)I-labeled antitumor antibodies. METHODS: As an example of this paradigm, quantitative image and kinetic analyses of anti-A33 humanized mAb (also known as "A33") were performed in 11 colorectal cancer patients. Serial whole-body PET scans of (124)I-labeled A33 and blood samples were acquired and the resulting tissue time-activity data for each patient were fit to a nonlinear compartmental model using the SAAM II computer code. RESULTS: Excellent agreement was observed between fitted and measured parameters of tumor uptake, "off-target" uptake in bowel mucosa, blood clearance, tumor antigen levels, and percent antigen occupancy. CONCLUSION: This approach should be generally applicable to antibody-antigen systems in human tumors for which the masses of antigen-expressing tumor and of normal tissues can be estimated and for which antibody kinetics can be measured with PET. Ultimately, based on each patient's resulting "best-fit" nonlinear model, a patient-specific optimum mAb dose (in micromoles, for example) may be derived.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias Colorrectales/diagnóstico por imagen , Neoplasias Colorrectales/tratamiento farmacológico , Modelos Biológicos , Terapia Molecular Dirigida , Tomografía de Emisión de Positrones , Medicina de Precisión , Animales , Anticuerpos Monoclonales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Radioisótopos de Yodo , Cinética , Ratones
4.
Proc Natl Acad Sci U S A ; 109(15): 5797-802, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22454499

RESUMEN

Recombinant poxviruses (vaccinia and fowlpox) expressing tumor-associated antigens are currently being evaluated in clinical trials as cancer vaccines to induce tumor-specific immune responses that will improve clinical outcome. To test whether a diversified prime and boost regimen targeting NY-ESO-1 will result in clinical benefit, we conducted two parallel phase II clinical trials of recombinant vaccinia-NY-ESO-1 (rV-NY-ESO-1), followed by booster vaccinations with recombinant fowlpox-NY-ESO-1 (rF-NY-ESO-1) in 25 melanoma and 22 epithelial ovarian cancer (EOC) patients with advanced disease who were at high risk for recurrence/progression. Integrated NY-ESO-1-specific antibody and CD4(+) and CD8(+) T cells were induced in a high proportion of melanoma and EOC patients. In melanoma patients, objective response rate [complete and partial response (CR+PR)] was 14%, mixed response was 5%, and disease stabilization was 52%, amounting to a clinical benefit rate (CBR) of 72% in melanoma patients. The median PFS in the melanoma patients was 9 mo (range, 0-84 mo) and the median OS was 48 mo (range, 3-106 mo). In EOC patients, the median PFS was 21 mo (95% CI, 16-29 mo), and median OS was 48 mo (CI, not estimable). CD8(+) T cells derived from vaccinated patients were shown to lyse NY-ESO-1-expressing tumor targets. These data provide preliminary evidence of clinically meaningful benefit for diversified prime and boost recombinant pox-viral-based vaccines in melanoma and ovarian cancer and support further evaluation of this approach in these patient populations.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vectores Genéticos/genética , Melanoma/inmunología , Proteínas de la Membrana/inmunología , Neoplasias Ováricas/inmunología , Vacunación , Vacunas Sintéticas/inmunología , Virus Vaccinia/genética , Formación de Anticuerpos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Virus de la Viruela de las Aves de Corral/genética , Humanos , Melanoma/patología , Neoplasias Ováricas/patología , Resultado del Tratamiento
5.
Int J Cancer ; 135(5): 1142-52, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24482145

RESUMEN

The prognosis of head and neck squamous cell carcinoma (HNSCC) patients remains poor. The identification of high-risk subgroups is needed for the development of custom-tailored therapies. The expression of cancer-testis antigens (CTAs) has been linked to a worse prognosis in other cancer types; however, their prognostic value in HNSCC is unclear because only few patients have been examined and data on CTA protein expression are sparse. A tissue microarray consisting of tumor samples from 453 HNSCC patients was evaluated for the expression of CTA proteins using immunohistochemistry. Frequency of expression and the subcellular expression pattern (nuclear, cytoplasmic, or both) was recorded. Protein expression of melanoma antigen (MAGE)-A family CTA, MAGE-C family CTA and NY-ESO-1 was found in approximately 30, 7 and 4% of tumors, respectively. The subcellular expression pattern in particular had a marked impact on the patients' prognosis. Median overall survival (OS) of patients with (i) simultaneous cytoplasmic and nuclear expression compared to (ii) either cytoplasmic or nuclear expression and (iii) negative patients was 23.0 versus 109.0 versus 102.5 months, for pan-MAGE (p < 0.0001), 46.6 versus 50.0 versus 109.0 for MAGE-A3/A4 (p = 0.0074) and 13.3 versus 50.0 versus 100.2 months for NY-ESO-1 (p = 0.0019). By multivariate analysis, these factors were confirmed as independent markers for poor survival. HNSCC patients showing protein expression of MAGE-A family members or NY-ESO-1 represent a subgroup with an extraordinarily poor survival. The development of immunotherapeutic strategies targeting these CTA may, therefore, be a promising approach to improve the outcome of HNSCC patients.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/inmunología , Biomarcadores de Tumor/biosíntesis , Carcinoma de Células Escamosas/mortalidad , Neoplasias de Cabeza y Cuello/mortalidad , Antígenos Específicos del Melanoma/biosíntesis , Proteínas de la Membrana/biosíntesis , Proteínas de Neoplasias/inmunología , Adulto , Anciano , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/radioterapia , Citoplasma/inmunología , Femenino , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/radioterapia , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/inmunología , Pronóstico , Riesgo , Carcinoma de Células Escamosas de Cabeza y Cuello
6.
Eur J Immunol ; 43(4): 989-1000, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23436617

RESUMEN

Cancer vaccines have yet to yield clinical benefit, despite the measurable induction of humoral and cellular immune responses. As immunosuppression by CD4(+) CD25(+) regulatory T (Treg) cells has been linked to the failure of cancer immunotherapy, blocking suppression is therefore critical for successful clinical strategies. Here, we addressed whether a lyophilized preparation of Streptococcus pyogenes (OK-432), which stimulates Toll-like receptors, could overcome Treg-cell suppression of CD4(+) T-cell responses in vitro and in vivo. OK-432 significantly enhanced in vitro proliferation of CD4(+) effector T cells by blocking Treg-cell suppression and this blocking effect depended on IL-12 derived from antigen-presenting cells. Direct administration of OK-432 into tumor-associated exudate fluids resulted in a reduction of the frequency and suppressive function of CD4(+) CD25(+) Foxp3(+) Treg cells. Furthermore, when OK-432 was used as an adjuvant of vaccination with HER2 and NY-ESO-1 for esophageal cancer patients, NY-ESO-1-specific CD4(+) T-cell precursors were activated, and NY-ESO-1-specific CD4(+) T cells were detected within the effector/memory T-cell population. CD4(+) T-cell clones from these patients had high-affinity TCRs and recognized naturally processed NY-ESO-1 protein presented by dendritic cells. OK-432 therefore inhibits Treg-cell function and contributes to the activation of high-avidity tumor antigen-specific naive T-cell precursors.


Asunto(s)
Terapia de Inmunosupresión , Streptococcus pyogenes/inmunología , Linfocitos T Reguladores/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Antígenos CD4/metabolismo , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Exudados y Transudados/inmunología , Humanos , Interleucina-12/farmacología , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Proteínas de la Membrana/administración & dosificación , Proteínas de la Membrana/inmunología , Neoplasias/inmunología , Picibanil/administración & dosificación , Picibanil/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo
7.
J Immunol ; 188(8): 3851-8, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22427632

RESUMEN

Tumor Ag-specific CD4(+) T cells play important functions in tumor immunosurveillance, and in certain cases they can directly recognize HLA class II-expressing tumor cells. However, the underlying mechanism of intracellular Ag presentation to CD4(+) T cells by tumor cells has not yet been well characterized. We analyzed two naturally occurring human CD4(+) T cell lines specific for different peptides from cytosolic tumor Ag NY-ESO-1. Whereas both lines had the same HLA restriction and a similar ability to recognize exogenous NY-ESO-1 protein, only one CD4(+) T cell line recognized NY-ESO-1(+) HLA class II-expressing melanoma cells. Modulation of Ag processing in melanoma cells using specific molecular inhibitors and small interfering RNA revealed a previously undescribed peptide-selective Ag-presentation pathway by HLA class II(+) melanoma cells. The presentation required both proteasome and endosomal protease-dependent processing mechanisms, as well as cytosolic heat shock protein 90-mediated chaperoning. Such tumor-specific pathway of endogenous HLA class II Ag presentation is expected to play an important role in immunosurveillance or immunosuppression mediated by various subsets of CD4(+) T cells at the tumor local site. Furthermore, targeted activation of tumor-recognizing CD4(+) T cells by vaccination or adoptive transfer could be a suitable strategy for enhancing the efficacy of tumor immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Epítopos de Linfocito T/inmunología , Proteínas HSP90 de Choque Térmico/inmunología , Péptidos/inmunología , Presentación de Antígeno , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/citología , Línea Celular Tumoral , Clonación Molecular , Citocinas/biosíntesis , Citocinas/inmunología , Citosol/inmunología , Citosol/metabolismo , Endosomas/genética , Endosomas/inmunología , Epítopos de Linfocito T/genética , Femenino , Proteínas HSP90 de Choque Térmico/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Activación de Linfocitos/inmunología , Melanoma/inmunología , Melanoma/patología , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Péptidos/genética , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , ARN Interferente Pequeño/genética
8.
Proc Natl Acad Sci U S A ; 108(49): 19695-700, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22114198

RESUMEN

One of the main challenges in cancer research is the development of vaccines that induce effective and long-lived protective immunity against tumors. Significant progress has been made in identifying members of the cancer testis antigen family as potential vaccine candidates. However, an ideal form for antigen delivery that induces robust and sustainable antigen-specific T-cell responses, and in particular of CD8(+) T lymphocytes, remains to be developed. Here we report the use of a recombinant nonpathogenic clone of Trypanosoma cruzi as a vaccine vector to induce vigorous and long-term T cell-mediated immunity. The rationale for using the highly attenuated T. cruzi clone was (i) the ability of the parasite to persist in host tissues and therefore to induce a long-term antigen-specific immune response; (ii) the existence of intrinsic parasite agonists for Toll-like receptors and consequent induction of highly polarized T helper cell type 1 responses; and (iii) the parasite replication in the host cell cytoplasm, leading to direct antigen presentation through the endogenous pathway and consequent induction of antigen-specific CD8(+) T cells. Importantly, we found that parasites expressing a cancer testis antigen (NY-ESO-1) were able to elicit human antigen-specific T-cell responses in vitro and solid protection against melanoma in a mouse model. Furthermore, in a therapeutic protocol, the parasites expressing NY-ESO-1 delayed the rate of tumor development in mice. We conclude that the T. cruzi vector is highly efficient in inducing T cell-mediated immunity and protection against cancer cells. More broadly, this strategy could be used to elicit a long-term T cell-mediated immunity and used for prophylaxis or therapy of chronic infectious diseases.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas de la Membrana/inmunología , Trypanosoma cruzi/inmunología , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Western Blotting , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/parasitología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/parasitología , Vacunas contra el Cáncer/administración & dosificación , Línea Celular Tumoral , Células Cultivadas , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/parasitología , Enfermedad de Chagas/prevención & control , Ensayo de Inmunoadsorción Enzimática , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Inmunización/métodos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Neoplasias Experimentales/prevención & control , Transfección/métodos , Trypanosoma cruzi/genética , Trypanosoma cruzi/metabolismo
9.
Proc Natl Acad Sci U S A ; 108(40): 16723-8, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21933959

RESUMEN

Ipilimumab, a monoclonal antibody against cytotoxic T lymphocyte antigen 4 (CTLA-4), has been shown to improve survival in patients with advanced metastatic melanoma. It also enhances immunity to NY-ESO-1, a cancer/testis antigen expressed in a subset of patients with melanoma. To characterize the association between immune response and clinical outcome, we first analyzed NY-ESO-1 serum antibody by ELISA in 144 ipilimumab-treated patients with melanoma and found 22 of 140 (16%) seropositive at baseline and 31 of 144 (22%) seropositive following treatment. These NY-ESO-1-seropositive patients had a greater likelihood of experiencing clinical benefit 24 wk after ipilimumab treatment than NY-ESO-1-seronegative patients (P = 0.02, relative risk = 1.8, two-tailed Fisher test). To understand why some patients with NY-ESO-1 antibody failed to experience clinical benefit, we analyzed NY-ESO-1-specific CD4(+) and CD8(+) T-cell responses by intracellular multicytokine staining in 20 NY-ESO-1-seropositive patients and found a surprising dissociation between NY-ESO-1 antibody and CD8 responses in some patients. NY-ESO-1-seropositive patients with associated CD8(+) T cells experienced more frequent clinical benefit (10 of 13; 77%) than those with undetectable CD8(+) T-cell response (one of seven; 14%; P = 0.02; relative risk = 5.4, two-tailed Fisher test), as well as a significant survival advantage (P = 0.01; hazard ratio = 0.2, time-dependent Cox model). Together, our data suggest that integrated NY-ESO-1 immune responses may have predictive value for ipilimumab treatment and argue for prospective studies in patients with established NY-ESO-1 immunity. The current findings provide a strong rationale for the clinical use of modulators of immunosuppression with concurrent approaches to favor tumor antigen-specific immune responses, such as vaccines or adoptive transfer, in patients with cancer.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Antígeno CTLA-4/inmunología , Melanoma/inmunología , Proteínas de la Membrana/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Inmunohistoquímica , Ipilimumab , Persona de Mediana Edad , Modelos de Riesgos Proporcionales , Análisis de Supervivencia
10.
J Exp Med ; 204(4): 831-40, 2007 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-17389240

RESUMEN

Specific targets of cellular immunity in human premalignancy are largely unknown. Monoclonal gammopathy of undetermined significance (MGUS) represents a precursor lesion to myeloma (MM). We show that antigenic targets of spontaneous immunity in MGUS differ from MM. MGUS patients frequently mount a humoral and cellular immune response against SOX2, a gene critical for self-renewal in embryonal stem cells. Intranuclear expression of SOX2 marks the clonogenic CD138(-) compartment in MGUS. SOX2 expression is also detected in a proportion of CD138(+) cells in MM patients. However, these patients lack anti-SOX2 immunity. Cellular immunity to SOX2 inhibits the clonogenic growth of MGUS cells in vitro. Detection of anti-SOX2 T cells predicts favorable clinical outcome in patients with asymptomatic plasmaproliferative disorders. Harnessing immunity to antigens expressed by tumor progenitor cells may be critical for prevention and therapy of human cancer.


Asunto(s)
Células Madre Embrionarias/inmunología , Proteínas HMGB/inmunología , Paraproteinemias/inmunología , Paraproteinemias/metabolismo , Factores de Transcripción/inmunología , Biomarcadores , Proliferación Celular , Células Cultivadas , Progresión de la Enfermedad , Proteínas HMGB/metabolismo , Salud , Humanos , Inmunoglobulina G/inmunología , Mieloma Múltiple/inmunología , Mieloma Múltiple/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Paraproteinemias/patología , Paraproteinemias/terapia , Factores de Transcripción SOXB1 , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Resultado del Tratamiento
11.
Mol Imaging ; 12(1): 67-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23348793

RESUMEN

Based on their inability to express argininosuccinate synthetase (ASS), some cancer entities feature the characteristic of L-arginine (Arg) auxotrophy. This inability to intrinsically generate Arg makes them applicable for arginine deiminase (ADI) treatment, an Arg-depleting drug. Arg is also used for the synthesis of endothelial nitric oxide (NO), which mainly confers vasodilatation but is also considered to have a major influence on tumor vascularization. The purpose of this study was to define changes in tumor vasculature in an ADI-treated melanoma xenograft mouse model using the blood pool agent AngioSense 750 and fluorescence molecular tomography (FMT). We used an ASS-negative melanoma xenograft mouse model and subjected it to weekly ADI treatment. Changes in tumor size were measured, and alterations in tumor vasculature were depicted by FMT and CD31 immunohistochemistry (IHC). On ADI treatment and effective antitumor therapy, we observed a drop in NO plasma levels and visualized changes in tumor vascularization with FMT and IHC. ADI treatment in melanoma xenografts has a tumor-reducing effect, which can be noninvasively imaged by quantifying tumor vascularization with FMT and IHC.


Asunto(s)
Hidrolasas/farmacología , Melanoma/irrigación sanguínea , Melanoma/tratamiento farmacológico , Imagen Óptica/métodos , Tomografía/métodos , Animales , Argininosuccinato Sintasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Inmunohistoquímica , Ratones , Ratones SCID , Imagen Molecular/métodos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Óxido Nítrico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Cancer Immun ; 13: 12, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23882157

RESUMEN

During cancer progression, malignant cells may evade immunosurveillance. However, evidence for immunological escape in humans is scarce. We report here the clinical course of a melanoma patient whose initial tumor was positive for the antigens NY-ESO-1, MAGE-C1, and Melan-A. Upon immunization with a recombinant vaccinia/fowlpox NY-ESO-1 construct, the patient experienced a mixed clinical response and spreading of the NY-ESO-1 epitopes in the CD4+ T cell compartment. After NY-ESO-1 protein + CpG immunization, the patient's anti-NY-ESO-1 IgG response increased. Over the following years, progressing lesions were resected and found to be NY-ESO-1-negative while being positive for MAGE-C1, Melan-A, and MHC-I. The fatal, inoperable brain metastasis was analyzed after his death and also proved to be NY-ESO-1-negative, while being positive for MAGE-C1 and Melan-A, as well as MHC-I. We propose that cancer control and cancer escape in this patient were governed by NY-ESO-1-specific immunological pressure. Our findings provide evidence for the existence of immunoediting and immunoescape in this cancer patient.


Asunto(s)
Antígenos de Neoplasias/inmunología , Melanoma/inmunología , Proteínas de la Membrana/inmunología , Humanos , Inmunohistoquímica , Masculino , Melanoma/patología , Melanoma/secundario , Persona de Mediana Edad
13.
Cancer Immun ; 13: 3, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23390374

RESUMEN

We investigated whether antibodies against intracellular tumor-associated antigens support tumor-specific immunity when administered together with a treatment that destroys the tumor. We propose that released antigens form immune complexes with the antibodies, which are then efficiently taken up by dendritic cells. We cloned the first human monoclonal antibodies against the Cancer/Testis (CT) antigen, NY-ESO-1. We tested whether the monoclonal anti-NY-ESO-1 antibody (12D7) facilitates cross-presentation of a NY-ESO-1-derived epitope by dendritic cells to human CD8+ T cells, and whether this results in the maturation of dendritic cells in vitro. We investigated the efficacy of 12D7 in combination with chemotherapy using BALB/c mice bearing syngeneic CT26 tumors that express intracellular NY-ESO-1. Human dendritic cells that were incubated with NY-ESO-1:12D7 immune complexes efficiently stimulated NY-ESO-1(157-165)/HLA-A2-specific human CD8+ T cells to produce interferon-γ, whereas NY-ESO-1 alone did not. Furthermore, the incubation of dendritic cells with NY-ESO-1:12D7 immune complexes resulted in the maturation of dendritic cells. Treatment of BALB/c mice that bear CT26/NY-ESO-1 tumors with 5-fluorouracil (5-FU) plus 12D7 was significantly more effective than chemotherapy alone. We propose systemic injection of monoclonal antibodies (mAbs) against tumor-associated antigens plus a treatment that promotes the local release of those antigens resulting in immune complex formation as a novel therapeutic modality for cancer.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Antígenos de Neoplasias/inmunología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Proteínas de la Membrana/inmunología , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Diferenciación Celular/efectos de los fármacos , Clonación Molecular , Reactividad Cruzada/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Mapeo Epitopo , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Humanos , Ratones , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/patología , Resultado del Tratamiento
14.
Protein Expr Purif ; 92(2): 171-82, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24045055

RESUMEN

Melan-A is a cancer testis antigen commonly found in melanoma, and has been shown to stimulate the body's immune response against cancerous cells. We have developed and executed a process utilizing current good manufacturing practices (cGMP) to produce the 6 times-His tagged protein in C41DE3 Escherichia coli for use in Phase I clinical trials. Approximately 11 g of purified Melan-A were produced from a 20 L fed-batch fermentation. Purification was achieved through a three column process utilizing immobilized metal affinity, anion exchange, and cation exchange chromatography with a buffer system optimized for low-solubility, high LPS binding capacity proteins. The host cell proteins, residual DNA, and endotoxin concentration were well below limits for a prescribed dose with a final purity level of 91%.


Asunto(s)
Vacunas contra el Cáncer , Histidina/metabolismo , Antígeno MART-1/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Investigación Biomédica , Química Farmacéutica , Cromatografía por Intercambio Iónico , Fermentación , Histidina/química , Histidina/genética , Antígeno MART-1/química , Antígeno MART-1/genética , Estabilidad Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Reproducibilidad de los Resultados
15.
J Immunol ; 186(2): 1218-27, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21149605

RESUMEN

Immunization of cancer patients with vaccines containing full-length tumor Ags aims to elicit specific Abs and both CD4(+) and CD8(+) T cells. Vaccination with protein Ags, however, often elicits only CD4(+) T cell responses without inducing Ag-specific CD8(+) T cells, as exogenous protein is primarily presented to CD4(+) T cells. Recent data revealed that Ab-mediated targeting of protein Ags to cell surface receptors on dendritic cells could enhance the induction of both CD4(+) and CD8(+) T cells. We investigated in this study if these observations were applicable to NY-ESO-1, a cancer-testis Ag widely used in clinical cancer vaccine trials. We generated two novel targeting proteins consisting of the full-length NY-ESO-1 fused to the C terminus of two human mAbs against the human mannose receptor and DEC-205, both internalizing molecules expressed on APC. These targeting proteins were evaluated for their ability to activate NY-ESO-1-specific human CD4(+) and CD8(+) T cells in vitro. Both targeted NY-ESO-1 proteins rapidly bound to their respective targets on APC. Whereas nontargeted and Ab-targeted NY-ESO-1 proteins similarly activated CD4(+) T cells, cross-presentation to CD8(+) T cells was only efficiently induced by targeted NY-ESO-1. In addition, both mannose receptor and DEC-205 targeting elicited specific CD4(+) and CD8(+) T cells from PBLs of cancer patients. Receptor-specific delivery of NY-ESO-1 to APC appears to be a promising vaccination strategy to efficiently generate integrated and broad Ag-specific immune responses against NY-ESO-1 in cancer patients.


Asunto(s)
Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Proteínas de la Membrana/inmunología , Receptores de Superficie Celular/metabolismo , Secuencia de Aminoácidos , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD/inmunología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular , Células Clonales , Reactividad Cruzada/genética , Reactividad Cruzada/inmunología , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/inmunología , Receptor de Manosa , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Unión Proteica/inmunología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo
16.
Proc Natl Acad Sci U S A ; 107(11): 5088-93, 2010 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-20194765

RESUMEN

Autoantibodies, a hallmark of both autoimmunity and cancer, represent an easily accessible surrogate for measuring adaptive immune responses to cancer. Sera can now be assayed for reactivity against thousands of proteins using microarrays, but there is no agreed-upon standard to analyze results. We developed a set of tailored quality control and normalization procedures based on ELISA validation to allow patient comparisons and determination of individual cutoffs for specificity and sensitivity. Sera from 60 patients with pancreatic cancer, 51 patients with ovarian cancer, and 53 age-matched healthy donors were used to assess the binding of IgG antibodies against a panel of >8000 human antigens using protein microarrays and fluorescence detection. The resulting data interpretation led to the definition and ranking of proteins with preferred recognition by the sera from cancer patients in comparison with healthy donors, both by frequency and strength of signal. We found that 202 proteins were preferentially immunogenic in ovarian cancer sera compared to 29 in pancreatic cancer, with few overlaps. Correlates of autoantibody signatures with known tumor expression of corresponding antigens, functional pathways, clinical stage, and outcome were examined. Serological analysis of arrays displaying the complete human proteome (seromics) represents a new era in cancer immunology, opening the way to defining the repertoire of the humoral immune response to cancer.


Asunto(s)
Neoplasias Ováricas/sangre , Neoplasias Pancreáticas/sangre , Análisis por Matrices de Proteínas/métodos , Proteoma/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Especificidad de Anticuerpos , Autoanticuerpos/inmunología , Biomarcadores de Tumor/inmunología , Donantes de Sangre , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Femenino , Genes Relacionados con las Neoplasias , Humanos , Persona de Mediana Edad , Proteínas de Neoplasias/sangre , Neoplasias Ováricas/genética , Neoplasias Ováricas/inmunología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Reproducibilidad de los Resultados
17.
Int J Cancer ; 130(7): 1598-606, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21547902

RESUMEN

Early detection of colorectal cancer (CRC) is currently based on fecal occult blood testing (FOBT) and colonoscopy, both which can significantly reduce CRC-related mortality. However, FOBT has low-sensitivity and specificity, whereas colonoscopy is labor- and cost-intensive. Therefore, the discovery of novel biomarkers that can be used for improved CRC screening, diagnosis, staging and as targets for novel therapies is of utmost importance. To identify novel CRC biomarkers we utilized representational difference analysis (RDA) and characterized a colon cancer associated transcript (CCAT1), demonstrating consistently strong expression in adenocarcinoma of the colon, while being largely undetectable in normal human tissues (p < 000.1). CCAT1 levels in CRC are on average 235-fold higher than those found in normal mucosa. Importantly, CCAT1 is strongly expressed in tissues representing the early phase of tumorigenesis: in adenomatous polyps and in tumor-proximal colonic epithelium, as well as in later stages of the disease (liver metastasis, for example). In CRC-associated lymph nodes, CCAT1 overexpression is detectable in all H&E positive, and 40.0% of H&E and immunohistochemistry negative lymph nodes, suggesting very high sensitivity. CCAT1 is also overexpressed in 40.0% of peripheral blood samples of patients with CRC but not in healthy controls. CCAT1 is therefore a highly specific and readily detectable marker for CRC and tumor-associated tissues.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias del Colon/genética , Precursores del ARN/genética , ARN Neoplásico/biosíntesis , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Pólipos Adenomatosos/diagnóstico , Pólipos Adenomatosos/genética , Adolescente , Secuencia de Aminoácidos , Secuencia de Bases , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Neoplasias del Colon/diagnóstico , Neoplasias del Colon/patología , Detección Precoz del Cáncer/métodos , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Ganglios Linfáticos/metabolismo , Datos de Secuencia Molecular , Membrana Mucosa/metabolismo , Metástasis de la Neoplasia , Precursores del ARN/análisis , ARN Neoplásico/genética , Sensibilidad y Especificidad , Regulación hacia Arriba
18.
Int J Cancer ; 130(3): 584-92, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21413013

RESUMEN

NY-ESO-1 is a prototypic cancer/testis antigen. In a recent phase I clinical trial, we vaccinated 13 patients bearing NY-ESO-1-expressing tumors with a complex of cholesterol-bearing hydrophobized pullulan (CHP) and NY-ESO-1 protein (CHP-NY-ESO-1) and showed efficient induction of NY-ESO-1 antibody, and CD4 and CD8 T cell responses using peripheral blood from the patients. In our study, we analyzed heteroclitic serological responses in those patients after vaccination. Serological response against 11 tumor antigens including MAGE-A1, MAGE-A3, MAGE-A4, CT7/MAGEC1, CT10/MAGEC2, CT45, CT46/HORMAD1, SOX2, SSX2, XAGE1B and p53 was examined by enzyme-linked immunosorbent assay (ELISA) using sera from ten vaccinated patients. Expression of tumor antigens was determined by reverse transcription-polymerase chain reaction or immunohistochemistry. Eight of nine patients who showed antibody responses against NY-ESO-1 also showed an antibody response against at least 1 of these 11 tumor antigens after vaccination. In one patient, seven tumor antigens were recognized. Specificity analysis of the antibody response by ELISA using control recombinant proteins and synthetic peptides and by Western blot showed that the response was not against His6-tag and/or bacterial products included in a preparation of CHP-NY-ESO-1 used for vaccination. Thus, heteroclitic serological responses appear to be indicative of the overall immune response against the tumor, and their analysis could be useful for immune monitoring in cancer vaccine.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Neoplasias Esofágicas/inmunología , Proteínas de la Membrana/inmunología , Neoplasias de la Próstata/inmunología , Animales , Anticuerpos Antineoplásicos/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular Tumoral , Neoplasias Esofágicas/sangre , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Próstata/sangre
19.
Proc Natl Acad Sci U S A ; 106(13): 5082-7, 2009 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-19289842

RESUMEN

Epidermal Growth Factor Receptor (EGFR) is involved in stimulating the growth of many human tumors, but the success of therapeutic agents has been limited in part by interference from the EGFR on normal tissues. Previously, we reported an antibody (mab806) against a truncated form of EGFR found commonly in gliomas. Remarkably, it also recognizes full-length EGFR on tumor cells but not on normal cells. However, the mechanism for this activity was unclear. Crystallographic structures for Fab:EGFR(287-302) complexes of mAb806 (and a second, related antibody, mAb175) show that this peptide epitope adopts conformations similar to those found in the wtEGFR. However, in both conformations observed for wtEGFR, tethered and untethered, antibody binding would be prohibited by significant steric clashes with the CR1 domain. Thus, these antibodies must recognize a cryptic epitope in EGFR. Structurally, it appeared that breaking the disulfide bond preceding the epitope might allow the CR1 domain to open up sufficiently for antibody binding. The EGFR(C271A/C283A) mutant not only binds mAb806, but binds with 1:1 stoichiometry, which is significantly greater than wtEGFR binding. Although mAb806 and mAb175 decrease tumor growth in xenografts displaying mutant, overexpressed, or autocrine stimulated EGFR, neither antibody inhibits the in vitro growth of cells expressing wtEGFR. In contrast, mAb806 completely inhibits the ligand-associated stimulation of cells expressing EGFR(C271A/C283A). Clearly, the binding of mAb806 and mAb175 to the wtEGFR requires the epitope to be exposed either during receptor activation, mutation, or overexpression. This mechanism suggests the possibility of generating antibodies to target other wild-type receptors on tumor cells.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antineoplásicos/inmunología , Receptores ErbB/inmunología , Proteínas de Neoplasias/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antineoplásicos/uso terapéutico , Complejo Antígeno-Anticuerpo/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Cristalografía por Rayos X , Epítopos , Humanos , Ratones , Ratones Desnudos , Conformación Proteica , Desnaturalización Proteica/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
J Immunol ; 183(7): 4800-8, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19734225

RESUMEN

Vaccination with cancer/testis Ag MAGE-A3 in the form of recombinant protein often induces specific humoral and cellular immune responses. Although Ag-specific CD4+ T cells following vaccination are detectable by cytokine production after a single in vitro stimulation, their detection before vaccination is difficult because of low frequency. In this study, we have applied a sensitive method using CD154 (CD40L) staining to detect MAGE-A3-specific CD4+ T cells. MAGE-A3-specific T cell responses were analyzed in four healthy donors, two lung cancer patients with spontaneous serum Abs to MAGE-A3, and two baseline seronegative lung cancer patients throughout vaccination with MAGE-A3 protein. MAGE-A3-specific CD4+ T cells were detected in all individuals tested, at low frequency in healthy donors and seronegative cancer patients and higher frequency in patients seropositive for MAGE-A3. Polyclonal expansion of CD154-expressing CD4+ T cells after cell sorting generated a large number of MAGE-A3-specific CD4+ T cell lines from all individuals tested, enabling full characterization of peptide specificity, HLA-restriction, and avidity. Application of this method to cancer patients vaccinated with MAGE-A3 protein with or without adjuvant revealed that protein vaccination induced oligoclonal activation of MAGE-A3-specific CD4+ T cells. It appeared that MAGE-A3 protein vaccination in the presence of adjuvant selectively expanded high avidity CD4+ T cells, whereas high avidity T cells disappeared after multiple vaccinations with MAGE-A3 protein alone.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Activación de Linfocitos/inmunología , Proteínas de Neoplasias/inmunología , Vacunas de Subunidad/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/patología , Vacunas contra el Cáncer/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Línea Celular Transformada , Técnicas de Cocultivo , Humanos , Inmunización Secundaria , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Factores de Tiempo , Células Tumorales Cultivadas , Vacunas de Subunidad/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA