Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Eur J Immunol ; 42(7): 1893-905, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22585674

RESUMEN

Surface expression of the IL-2 receptor α-chain (CD25) has been used to discriminate between CD4(+) CD25(HI) FOXP3(+) regulatory T (Treg) cells and CD4(+) CD25(NEG) FOXP3(-) non-Treg cells. However, this study reports that the majority of resting human memory CD4(+) FOXP3(-) T cells expresses intermediate levels of CD25 and that CD25 expression can be used to delineate a functionally distinct memory subpopulation. The CD25(NEG) memory T-cell population contains the vast majority of late differentiated cells that respond to antigens associated with chronic immune responses and are increased in patients with systemic lupus erythematosus (SLE). In contrast, the CD25(INT) memory T cells respond to antigens associated with recall responses, produce a greater array of cytokines, and are less dependent on costimulation for effector responses due to their expression of CD25. Lastly, compared to the CD25(NEG) and Treg-cell populations, the CD25(INT) memory population is lost to a greater degree from the blood of cancer patients treated with IL-2. Collectively, these results show that in humans, a large proportion of CD4(+) memory T cells express intermediate levels of CD25, and this CD25(INT) FOXP3(-) subset is a functionally distinct memory population that is uniquely affected by IL-2.


Asunto(s)
Factores de Transcripción Forkhead/inmunología , Memoria Inmunológica/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Neoplasias Renales/inmunología , Lupus Eritematoso Sistémico/inmunología , Melanoma/inmunología , Linfocitos T Reguladores/inmunología , Adulto , Femenino , Citometría de Flujo , Humanos , Inmunofenotipificación/métodos , Interleucina-2/uso terapéutico , Neoplasias Renales/sangre , Neoplasias Renales/tratamiento farmacológico , Lupus Eritematoso Sistémico/sangre , Masculino , Melanoma/sangre , Melanoma/tratamiento farmacológico , Persona de Mediana Edad , Adulto Joven
2.
Eur J Immunol ; 40(5): 1375-85, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20186876

RESUMEN

Lymphodeleption prior to adoptive transfer of tumor-specific T cells greatly improves the clinical efficacy of adoptive T-cell therapy for patients with advanced melanoma, and increases the therapeutic efficacy of cancer vaccines in animal models. Lymphodepletion reduces competition between lymphocytes, and thus creates "space" for enhanced expansion and survival of tumor-specific T cells. Within the lymphodepleted host, Ag-specific T cells still need to compete with other lymphocytes that undergo lymphopenia-driven proliferation. Herein, we describe the relative capacity of naïve T cells, Treg, and NK cells to undergo lymphopenia-driven proliferation. We found that the major population that underwent lymphopenia-driven proliferation was the CD122+ memory-like T-cell population (CD122+CD8+ Treg), and these cells competed with Ag-driven proliferation of melanoma-specific T cells. Removal of CD122+CD8+ Treg resulted in a greater expansion of tumor-specific T cells and tumor infiltration of functional effector/memory T cells. Our results demonstrate the lymphopenia-driven proliferation of CD122+CD8+ Treg in reconstituted lymphodepleted mice limited the antitumor efficacy of DC vaccination in conjunction with adoptive transfer of tumor-specific T cells.


Asunto(s)
Linfocitos T CD8-positivos/trasplante , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunoterapia Adoptiva , Depleción Linfocítica , Linfopenia/inmunología , Melanoma Experimental/terapia , Linfocitos T Reguladores/inmunología , Animales , Antígenos CD8/análisis , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , División Celular , Células Dendríticas/trasplante , Memoria Inmunológica , Interleucina-15/deficiencia , Subunidad beta del Receptor de Interleucina-2/análisis , Interleucina-7/farmacología , Interleucina-7/uso terapéutico , Células Asesinas Naturales/citología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Noqueados , Quimera por Radiación , Bazo/citología , Linfocitos T Reguladores/citología
3.
J Immunol ; 183(6): 3682-9, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19692636

RESUMEN

Regulatory T (Treg) cells represent a major roadblock to the induction of antitumor immunity through vaccine approaches. TGF-beta is a cytokine implicated in the generation and maintenance of Treg cells, as well as in their suppressive function. These experiments examined whether the generation of tumor-sensitized Treg cells was TGF-beta dependent and evaluated whether TGF-beta produced by Treg cells blocked the priming of tumor-specific T cells in vaccinated reconstituted lymphopenic mice. We show that tumor-sensitized Treg cells (CD25(+)/FoxP3(+)) obtained from tumor-bearing mice block the generation of tumor-specific T cells in reconstituted lymphopenic mice. Strikingly, this suppression is absent if tumor-sensitized Treg cells are acquired from tumor-bearing mice expressing the dominant-negative TGFbetaRII in T cells. This loss of suppression was a result of the crucial role of TGF-beta in generating tumor-sensitized Treg cells, and not due to the insensitivity of naive or tumor-primed effector T cells to the direct suppressive influence of TGF-beta. We conclude that blocking TGF-beta in a tumor-bearing host can inhibit the induction of highly suppressive tumor-sensitized Treg cells. These data suggest that an integrative strategy combining "up-front" Treg cell ablation followed by vaccination and TGF-beta blockade may limit generation of new tumor-sensitized Treg cells and improve the generation of therapeutic immune responses in patients with cancer.


Asunto(s)
Neoplasias Experimentales/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Células Cultivadas , Inmunidad , Inmunoterapia Adoptiva/métodos , Ratones , Ratones Noqueados , Transducción de Señal/inmunología
4.
Eur J Immunol ; 39(11): 3121-33, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19839008

RESUMEN

We reported previously that vaccination of reconstituted, lymphopenic mice resulted in a higher frequency of tumor-specific effector T cells with therapeutic activity than vaccination of normal mice. Here, we show that lymphopenic mice reconstituted with spleen cells from tumor-bearing mice (TBM), a situation that resembles the clinical condition, failed to generate tumor-specific T cells with therapeutic efficacy. However, depletion of CD25(+) Treg from the spleen cells of TBM restored tumor-specific priming and therapeutic efficacy. Adding back TBM CD25(+) Treg to CD25(-) naïve and TBM donor T cells prior to reconstitution confirmed their suppressive role. CD25(+) Treg from TBM prevented priming of tumor-specific T cells since subsequent depletion of CD4(+) T cells did not restore therapeutic efficacy. This effect may not be antigen-specific as three histologically distinct tumors generated CD25(+) Treg that could suppress the T-cell immune response to a melanoma vaccine. Importantly, since ex vivo depletion of CD25(+) Treg from TBM spleen cells prior to reconstitution and vaccination fully restored the generation of therapeutic effector T cells, even in animals with established tumor burden, we have initiated a translational clinical trial of this strategy in patients with metastatic melanoma.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Linfopenia/inmunología , Melanoma Experimental/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Subunidad alfa del Receptor de Interleucina-2/inmunología , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
J Transl Med ; 8: 106, 2010 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-21034498

RESUMEN

BACKGROUND: The complex data sets generated by higher-order polychromatic flow cytometry experiments are a challenge to analyze. Here we describe Exhaustive Expansion, a data analysis approach for deriving hundreds to thousands of cell phenotypes from raw data, and for interrogating these phenotypes to identify populations of biological interest given the experimental context. METHODS: We apply this approach to two studies, illustrating its broad applicability. The first examines the longitudinal changes in circulating human memory T cell populations within individual patients in response to a melanoma peptide (gp100209-2M) cancer vaccine, using 5 monoclonal antibodies (mAbs) to delineate subpopulations of viable, gp100-specific, CD8+ T cells. The second study measures the mobilization of stem cells in porcine bone marrow that may be associated with wound healing, and uses 5 different staining panels consisting of 8 mAbs each. RESULTS: In the first study, our analysis suggests that the cell surface markers CD45RA, CD27 and CD28, commonly used in historical lower order (2-4 color) flow cytometry analysis to distinguish memory from naïve and effector T cells, may not be obligate parameters in defining central memory T cells (TCM). In the second study, we identify novel phenotypes such as CD29+CD31+CD56+CXCR4+CD90+Sca1-CD44+, which may characterize progenitor cells that are significantly increased in wounded animals as compared to controls. CONCLUSIONS: Taken together, these results demonstrate that Exhaustive Expansion supports thorough interrogation of complex higher-order flow cytometry data sets and aids in the identification of potentially clinically relevant findings.


Asunto(s)
Citometría de Flujo/métodos , Anticuerpos Monoclonales/inmunología , Antígenos CD/inmunología , Humanos , Memoria Inmunológica , Linfocitos T/inmunología
6.
Clin Cancer Res ; 15(7): 2541-51, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19318471

RESUMEN

PURPOSE: The aim of this study was to characterize the primary gp100(209-2M)-specific T-cell response in vaccine-draining, metastases-free lymph nodes and peripheral blood of peptide-vaccinated stage I to III melanoma patients. EXPERIMENTAL DESIGN: After two or three gp100(209-2M) vaccinations, sentinel lymph nodes that drained both the primary tumor and adjacent vaccine sites were excised concomitant with wide excision of the tumor. Comparative 7-color flow cytometry phenotype analysis was done on gp100 tetramer-positive CD8(+) T cells from sentinel lymph nodes, closely proximate time-related peripheral blood mononuclear cells (PBMC) collected 2 to 4 weeks after sentinel lymph node excision, and on PBMC collected 6 months later after 7 or 11 more immunizations. Lymph node and peripheral blood T cells were tested for proliferative response, functional avidity, and tumor cell-induced CD107 mobilization. RESULTS: The frequencies of gp100-specific CD8(+) T cells from time-related PBMC and sentinel lymph nodes were comparable and were similar to those reported for virus-specific memory T cells. Their respective in vitro proliferation responses were also equivalent but statistically higher than proliferation responses of peripheral blood T cells collected after completion of the entire vaccine regimen. By contrast, functional avidity and CD107 responses were significantly higher in circulating T cells. Sentinel lymph node-derived, gp100-specific CD8(+) T cells predominantly expressed central and effector memory phenotype signatures, whereas there were higher frequencies of effector T cells in the peripheral blood. CONCLUSION: Priming immunization with gp100(209-2M) without coadministration of CD4(+) helper T cell-restricted antigens induced the effective expansion of peptide-specific central and effector memory CD8(+) T cells with high proliferation potential in vaccine-draining lymph nodes of stage I to III melanoma patients. Lymph node memory T cells gave rise to circulating gp100-specific effector T cells exhibiting increased functional maturation.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Melanoma/inmunología , Glicoproteínas de Membrana/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Memoria Inmunológica , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Proteínas de Membrana de los Lisosomas/análisis , Melanoma/patología , Biopsia del Ganglio Linfático Centinela , Antígeno gp100 del Melanoma
7.
Clin Cancer Res ; 14(16): 5270-83, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18698047

RESUMEN

PURPOSE: Effective cancer vaccines must both drive a strong CTL response and sustain long-term memory T cells capable of rapid recall responses to tumor antigens. We sought to characterize the phenotype and function of gp100 peptide-specific memory CD8+ T cells in melanoma patients after primary gp100(209-2M) immunization and assess the anamnestic response to boosting immunization. EXPERIMENTAL DESIGN: Eight-color flow cytometry analysis of gp100-specific CD8+ T cells was done on peripheral blood mononuclear cells collected shortly after the primary vaccine regimen, 12 to 24 months after primary vaccination, and after boosting immunization. The anamnestic response was assessed by comparing the frequency of circulating gp100-specific T cells before and after boosting. Gp100 peptide-induced in vitro functional avidity and proliferation responses and melanoma-stimulated T-cell CD107 mobilization were compared for cells from all three time points for multiple patients. RESULTS: The frequency of circulating gp100-specific memory CD8+ T cells was comparable with cytomegalovirus-specific and FLU-specific T cells in the same patients, and the cells exhibited anamnestic proliferation after boosting. Their phenotypes were not unique, and individual patients exhibited one of two distinct phenotype signatures that were homologous to either cytomegalovirus-specific or FLU-specific memory T cells. Gp100-specific memory T cells showed some properties of competent memory T cells, such as heightened in vitro peptide-stimulated proliferation and increase in central memory (TCM) differentiation when compared with T-cell responses measured after the primary vaccine regimen. However, they did not acquire enhanced functional avidity usually associated with competent memory T-cell maturation. CONCLUSIONS: Although vaccination with class I-restricted melanoma peptides alone can break tolerance to self-tumor antigens, it did not induce fully competent memory CD8+ T cells--even in disease-free patients. Data presented suggest other vaccine strategies will be required to induce functionally robust long-term memory T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Inmunización Secundaria , Melanoma/inmunología , Glicoproteínas de Membrana/inmunología , Neoplasias Cutáneas/inmunología , Vacunas contra el Cáncer/uso terapéutico , Citometría de Flujo , Humanos , Memoria Inmunológica , Melanoma/terapia , Fenotipo , Neoplasias Cutáneas/terapia , Antígeno gp100 del Melanoma
8.
J Transl Med ; 4: 24, 2006 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-16772043

RESUMEN

BACKGROUND: In vivo studies have recently demonstrated that interleukin 21 (IL-21) enhances the anti-tumor function of T-cells and NK cells in murine tumor models, and the combined use of IL-21 and IL-15 has resulted in prolonged tumor regression and survival in mice with previously established tumors. However, the combined anti-tumor effects of IL-21 and low dose IL-2 have not been studied even though IL-2 has been approved for human use, and, at low dose administration, stimulates the proliferation of memory T cells, and does not significantly increase antigen-induced apoptosis or regulatory T cell (Treg) expansion. This study examined whether recombinant IL-21 alone or in combination with low-dose IL-2 could improve the in vivo anti-tumor function of naïve, tumor-antigen specific CD8+ T cells in a gp100(25-33) T cell receptor transgenic pmel murine melanoma model. METHODS: Congenic C57BL/6 (Ly5.2) mice bearing subcutaneous B16F10 melanoma tumors were sublethally irradiated to induce lymphopenia. After irradiation naive pmel splenocytes were adoptively transferred, and mice were immunized with bone marrow-derived dendritic cells pulsed with human gp100(25-33) (hgp100(25-33)). Seven days after vaccination groups of mice received 5 consecutive days of intraperitoneal administration of IL-2 alone (20 x 10(3) IU), IL-21 alone (20 microg) or IL-21 and IL-2. Control animals received no cytokine therapy. RESULTS: IL-21 alone and IL-2 alone both delayed tumor progression, but only IL-21 significantly augmented long-term survival (20%) compared to the control group. However, combination therapy with IL-21 and IL-2 resulted in the highest long-term (>150 days) tumor-free survival frequency of 46%. Animals that were tumor-free for > 150 days demonstrated tumor-specific protection after rechallenge with B16F10 melanoma cells. At peak expansion (21 days post vaccination), the combination of IL-21 plus IL-2 resulted in a 2- to 3-fold higher absolute number of circulating tumor antigen-specific pmel CD8+ T cells than was stimulated by IL-2 or IL-21 alone. Pmel CD8+ T cells were predominantly partitioned into central memory (CD62L+/CD127+) or effector-memory (CD62L-/CD127+) phenotypes by day 28-post vaccination in IL-21 + IL-2 treated mice. CONCLUSION: These observations support the potential use of IL-21 and low-dose IL-2 therapy in combination with a tumor-antigen vaccine and lymphopenic conditioning in future cancer clinical trials to maintain high numbers of anti-tumor memory CD8+ T cells with the potential to sustain long term tumor regression and survival.

9.
J Clin Oncol ; 21(8): 1562-73, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12697882

RESUMEN

PURPOSE: To measure the CD8+ T-cell response to a melanoma peptide vaccine and to compare an every-2-weeks with an every-3-weeks vaccination schedule. PATIENTS AND METHODS: Thirty HLA-A2-positive patients with resected stage I to III melanoma were randomly assigned to receive vaccinations every 2 weeks (13 vaccines) or every 3 weeks (nine vaccines) for 6 months. The synthetic, modified gp100 peptide, g209-2M, and a control peptide, HPV16 E7, were mixed in incomplete Freund's adjuvant and injected subcutaneously. Peripheral blood mononuclear cells obtained before and after vaccination by leukapheresis were analyzed using a fluorescence-based HLA/peptide-tetramer binding assay and cytokine flow cytometry. RESULTS: Vaccination induced an increase in peptide-specific T cells in 28 of 29 patients. The median frequency of CD8+ T cells specific for the g209-2M peptide increased markedly from 0.02% before to 0.34% after vaccination (P <.0001). Eight patients (28%) exhibited peptide-specific CD8+ T-cell frequencies greater than 1%, including two patients with frequencies of 4.96% and 8.86%, respectively. Interferon alfa-2b-treated patients also had significant increases in tetramer-binding cells (P <.0001). No difference was observed between the every-2-weeks and the every-3-weeks vaccination schedules (P =.59). CONCLUSION: Flow cytometric analysis of HLA/peptide-tetramer binding cells was a reliable means of quantifying the CD8+ T-cell response to peptide immunization. This assay may be suitable for use in future trials to optimize different vaccination strategies. Concurrent interferon treatment did not inhibit the development of a peptide-specific immune response and vaccination every 2 weeks, and every 3 weeks produced similar results.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Antígeno HLA-A2/análisis , Melanoma/inmunología , Glicoproteínas de Membrana/administración & dosificación , Proteínas de Neoplasias/administración & dosificación , Neoplasias Cutáneas/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Citometría de Flujo , Fluorescencia , Adyuvante de Freund/administración & dosificación , Humanos , Interferón gamma/metabolismo , Leucaféresis , Masculino , Glicoproteínas de Membrana/efectos adversos , Persona de Mediana Edad , Proteínas de Neoplasias/efectos adversos , Péptidos , Proyectos Piloto , Resultado del Tratamiento , Antígeno gp100 del Melanoma
10.
Clin Cancer Res ; 10(2): 668-80, 2004 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-14760090

RESUMEN

Thirty-five HLA-A2(+) patients with completely resected stage I-III melanoma were vaccinated multiple times over 6 months with a modified melanoma peptide, gp100(209-2M), emulsified in Montanide adjuvant. Direct ex vivo gp100(209-2M) tetramer analysis of pre- and postvaccine peripheral blood mononuclear cells (PBMCs) demonstrated significant increases in the frequency of tetramer(+) CD8(+) T cells after immunization for 33 of 35 evaluable patients (median, 0.36%; range, 0.05-8.9%). Ex vivo IFN-gamma cytokine flow cytometry analysis of postvaccine PBMCs after brief gp100(209-2M) in vitro activation showed that for all of the patients studied tetramer(+) CD8(+) T cells produced IFN-gamma; however, some patients had significant numbers of tetramer(+) IFN-gamma(-) CD8(+)T cells suggesting functional anergy. Additionally, 8 day gp100(209-2M) in vitro stimulation (IVS) of pre- and postvaccine PBMCs resulted in significant expansion of tetramer(+) CD8(+) T cells from postvaccine cells for 34 patients, and these IVS tetramer(+) CD8(+) T cells were functionally responsive by IFN-gamma cytokine flow cytometry analysis after restimulation with either native or modified gp100 peptide. However, correlated functional and phenotype analysis of IVS-expanded postvaccine CD8(+) T cells demonstrated the proliferation of functionally anergic gp100(209-2M)- tetramer(+) CD8(+) T cells in several patients and also indicated interpatient variability of gp100(209-2M) stimulated T-cell proliferation. Flow cytometry analysis of cryopreserved postvaccine PBMCs from representative patients showed that the majority of tetramer(+) CD8+ T cells (78.1 +/- 4.2%) had either an "effector" (CD45 RA(+)/CCR7(-)) or an "effector-memory" phenotype (CD45RA(-)/CCR7(-)). Notably, analysis of PBMCs collected 12-24 months after vaccine therapy demonstrated the durable presence of gp100(209-2M)-specific memory CD8(+) T cells with high proliferation potential. Overall, this report demonstrates that after vaccination with a MHC class I-restricted melanoma peptide, resected nonmetastatic melanoma patients can mount a significant antigen-specific CD8(+) T-cell immune response with a functionally intact memory component. The data further support the combined use of tetramer binding and functional assays in correlated ex vivo and IVS settings as a standard for immunomonitoring of cancer vaccine patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer , Antígeno HLA-A2/metabolismo , Melanoma/inmunología , Melanoma/terapia , Glicoproteínas de Membrana/química , Proteínas de Neoplasias/química , Fragmentos de Péptidos/química , Antígenos de Neoplasias , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Citocinas/biosíntesis , Células Dendríticas/citología , Dimerización , Citometría de Flujo , Humanos , Memoria Inmunológica , Interferón gamma/metabolismo , Leucocitos Mononucleares/metabolismo , Melanoma/patología , Glicoproteínas de Membrana/inmunología , Metástasis de la Neoplasia , Proteínas de Neoplasias/inmunología , Células Neoplásicas Circulantes , Fragmentos de Péptidos/inmunología , Péptidos/química , Fenotipo , Linfocitos T/inmunología , Antígeno gp100 del Melanoma
11.
Int Rev Immunol ; 22(3-4): 283-319, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12745643

RESUMEN

Clinical evaluation of therapeutic tumor vaccines has resulted in examination and comparison of the types of immune function assays required to monitor tumor antigen-stimulated T cell effector function in immunized patients. Three of the most commonly used assays include ELISPOT, tetramer assay, and cytokine flow cytometry (CFC). Discussed are the method and principles for each assay and an assessment of important methodological, reagent, and data acquisition issues that are relevant for the accurate and effective use of the assays. The sensitivity and utility of the assays and present arguments advocating their integrated use in future immunomonitoring studies are also discussed.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Neoplasias/terapia , Vacunas contra el Cáncer/inmunología , Citocinas/análisis , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Monitoreo Fisiológico , Neoplasias/inmunología , Sensibilidad y Especificidad
12.
Immunotherapy ; 2(6): 799-816, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21091113

RESUMEN

Measuring cytokine production is an integral part of measuring immune response during immunotherapy. Current technologies allow the simultaneous quantification of multiple cytokines in a variety of tissues. Patterns of cytokine response can be referred to as cytokine profiles. This article discusses the experimental design and data analysis of a number of studies that examined cytokine profiles in humans. We highlight potential sources of variability, both due to assay nuances and the diversity of human populations. We present strategies for analyzing data, emphasizing both multidimensional analysis and the value of treating each donor as his or her own control.


Asunto(s)
Citocinas/biosíntesis , Inmunoterapia/métodos , Modelos Estadísticos , Proyectos de Investigación , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Estudios de Cohortes , Citocinas/genética , Citometría de Flujo/métodos , Perfilación de la Expresión Génica , Humanos , Inmunoensayo/métodos , Persona de Mediana Edad , Neoplasias/terapia
13.
Semin Oncol ; 37(5): 524-32, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21074068

RESUMEN

The existence of tumor-specific T cells, as well as their ability to be primed in cancer patients, confirms that the immune response can be deployed to combat cancer. However, there are obstacles that must be overcome to convert the ineffective immune response commonly found in the tumor environment to one that leads to sustained destruction of tumor. Members of the tumor necrosis factor (TNF) superfamily direct diverse immune functions. OX40 and its ligand, OX40L, are key TNF members that augment T-cell expansion, cytokine production, and survival. OX40 signaling also controls regulatory T-cell differentiation and suppressive function. Studies over the past decade have demonstrated that OX40 agonists enhance antitumor immunity in preclinical models using immunogenic tumors; however, treatment of poorly immunogenic tumors has been less successful. Combining strategies that prime tumor-specific T cells together with OX40 signaling could generate and maintain a therapeutic antitumor immune response.


Asunto(s)
Inmunoterapia/métodos , Neoplasias/inmunología , Neoplasias/terapia , Ligando OX40/inmunología , Receptores OX40/inmunología , Transducción de Señal/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Vacunas contra el Cáncer , Terapia Combinada , Humanos , Inmunoterapia Adoptiva , Ratones , Ligando OX40/agonistas , Receptores OX40/agonistas , Linfocitos T Reguladores/inmunología
14.
Curr Mol Med ; 9(6): 673-82, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19689294

RESUMEN

Since multiple lines of experimental and clinical data clearly identified regulatory T cells as an integral part of the immune response, these cells have become a major focus of investigation in tumor immunology. Regulatory T cells are in place to dampen ongoing immune responses and to prevent autoimmunity, but they also have profound effects in blocking therapeutic anti-tumor activity. Therefore regulatory T cells are seen as a major hurdle that must be overcome in order for cancer immunotherapy to reach its therapeutic potential. Regulatory T cells are heterogeneous with sub-populations that exhibit distinct functional features. Here we will review the individual sub-populations in regards to their mode of action and their potential impact on blocking anti-tumor immunity. Approaches to measure function and frequency of regulatory T cells in model systems and clinical trails will be discussed. Finally, we will describe possible ways to interfere with regulatory T cell-mediated immune suppression with the focus on recent pre-clinical and clinical findings.


Asunto(s)
Tolerancia Inmunológica , Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Linfocitos T Reguladores/inmunología , Animales , Vacunas contra el Cáncer/inmunología , Ensayos Clínicos como Asunto , Humanos
15.
J Immunother ; 31(1): 43-51, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18157011

RESUMEN

Recent developments in immunology have provided new strategies to induce and augment the immune response to cancer. Nonetheless, objective clinical responses after vaccination are rare and even when high frequencies of tumor-specific T cells are achieved after adoptive immunotherapy, tumor cells continue to evade the immune response. We hypothesize that 1 mechanism of resistance of tumor cells to destruction by T cells is an elevated threshold for the induction of apoptosis. Inhibitor of apoptosis proteins (IAPs) are overexpressed in various tumors and have been associated with treatment failure and poor prognosis. As the mitochondrial peptide second mitochondria-derived activator of caspase (Smac) can antagonize IAPs, we designed a GFP-Smac fusion protein with a granzyme B (GrB) cleavage site. This fusion protein should be cleaved when tumor-specific cytolytic T cells recognize the tumor and, using the pore-forming protein perforin, insert GrB into the target. Here we report that transfer of a construct encoding a novel eGFP-Smac fusion protein (pro-Smac) containing a specific cleavage site for GrB, into the poorly immunogenic mouse melanoma cell line, B16BL6-D5 (D5), sensitizes tumor cells for killing by tumor-specific wild type, but not perforin-deficient (perforin-knockout), effector T cells in vitro and in vivo. These results describe the first example of a tumor-specific, T-cell-mediated approach to amplify the GrB-mediated cytotoxicity pathway with a pro-Smac fusion protein and provide an innovative approach to overcome IAPs and improve the efficacy of immunotherapy.


Asunto(s)
Proteínas Portadoras/metabolismo , Inmunoterapia Adoptiva/métodos , Melanoma Experimental/terapia , Proteínas Mitocondriales/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Western Blotting , Proteínas Portadoras/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Femenino , Citometría de Flujo , Granzimas/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteínas Citotóxicas Formadoras de Poros/genética , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteínas Recombinantes de Fusión/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/trasplante , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA