Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
PLoS One ; 8(1): e54804, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23382974

RESUMEN

Strategically-paired Toll-like receptor (TLR) ligands induce a unique dendritic cell (DC) phenotype that polarizes Th1 responses. We therefore investigated pairing single TLR ligands with a non TLR-mediated danger signal to cooperatively induce distinct DC properties from cultured human monocytes. Adenosine triphosphate (ATP) and the TLR2 ligand lipoteichoic acid (LTA) selectively and synergistically induced expression of IL-23 and IL-1ß from cultured monocytes as determined by ELISA assays. Flow cytometric analysis revealed that a sizable sub-population of treated cells acquired DC-like properties including activated surface phenotype with trans-well assays showing enhanced migration towards CCR7 ligands. Such activated cells also preferentially deviated, in an IL-23 and IL-1-dependent manner, CD4(pos) T lymphocyte responses toward the IL-22(hi), IL-17(hi)/IFN-γ(lo) Th17 phenotype in standard in vitro allogeneic sensitization assays. Although pharmacological activation of either ionotropic or cAMP-dependent pathways acted in synergy with LTA to enhance IL-23, only inhibition of the cAMP-dependent pathway antagonized ATP-enhanced cytokine production. ATP plus atypical lipopolysaccharide from P. gingivalis (signaling through TLR2) was slightly superior to E. coli-derived LPS (TLR4 ligand) for inducing the high IL-23-secreting DC-like phenotype, but greatly inferior for inducing IL-12 p70 production when paired with IFN-γ, a distinction reflected in activated DCs' ability to deviate lymphocytes toward Th1. Collectively, our data suggest TLR2 ligands encountered by innate immune cells in an environment with physiologically-relevant levels of extracellular ATP can induce a distinct activation state favoring IL-23- and IL-1ß-dependent Th17 type response.


Asunto(s)
Activación de Linfocitos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Células Th17/inmunología , Células Th17/metabolismo , Receptor Toll-Like 2/metabolismo , Adenosina Trifosfato/farmacología , Adenilil Ciclasas/metabolismo , Células Cultivadas , Quimiotaxis/efectos de los fármacos , Quimiotaxis/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Sinergismo Farmacológico , Humanos , Mediadores de Inflamación , Lipopolisacáridos/farmacología , Prueba de Cultivo Mixto de Linfocitos , Monocitos/efectos de los fármacos , Fenotipo , Subunidades de Proteína , Transducción de Señal , Ácidos Teicoicos/farmacología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 4/metabolismo
2.
J Surg Oncol ; 103(6): 550-4, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21480248

RESUMEN

Regional draining lymph nodes (LNs) play a pivotal role in initiating immune responses. However, the presence of metastases may compromise their normal immunological function. Preclinical studies indicate that despite metastases, early tumor-draining LNs are still a rich source of sensitized T cells. Recently, we found that dendritic (DC)-tumor fusion hybrids were capable of stimulating therapeutic T-cell generation in the LN. However, this response is regulated by a tumor-specific suppression mechanism(s). Reversal of these dysfunctions would help the success of immunotherapy.


Asunto(s)
Traslado Adoptivo , Vacunas contra el Cáncer , Células Dendríticas/inmunología , Células Híbridas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Antígenos de Neoplasias/inmunología , Humanos , Terapia de Inmunosupresión , Ganglios Linfáticos/inmunología , Ratones , Subgrupos de Linfocitos T/metabolismo
3.
Hum Immunol ; 72(1): 24-31, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20951755

RESUMEN

It was originally reported that only a small fraction of total matured dendritic cells (DCs) produced interleukin (IL)-12, but it has never been determined whether different combinations of activating signals now shown to maximize secreted IL-12 do so through increasing output by the same IL-12 producers, or by recruiting additional cytokine-secreting cells. We therefore tested all combinations of bacterial lipopolysaccharide (LPS) (TLR4 ligand), R848 (TLR8 ligand), interferon (IFN)-γ, and CD40L for activating human monocyte-derived dendritic cells (DC), and determined by intracellular flow cytometry that enhanced IL-12 secretion was accomplished in large part by markedly increasing the proportion of cells producing IL-12, with the triple and quadruple combinations recruiting the most DC. This optimization requirement for multiple signals was not reflected in differential Toll-like receptor (TLR) expression by the cells. Interestingly, DCs activated with single TLR ligands plus IFN-γ were capable of responding with a second burst of IL-12 upon later CD40L stimulation, whereas DCs activated with R848 plus LPS were not, despite the trend of the latter for superior polarization of naive T cells toward IFN-γ-secreting Th1. These results have implications for the biology of IL-12-secreting DCs and choice of activation regimen for prospective use in DC-based immunotherapy.


Asunto(s)
Polaridad Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Células TH1/citología , Células TH1/inmunología , Antígenos de Superficie/inmunología , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Humanos , Interferón-alfa/inmunología , Interferón-alfa/metabolismo , Interleucina-12/inmunología , Interleucina-12/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/metabolismo , Fenotipo , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
4.
Head Neck ; 32(9): 1209-16, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20054852

RESUMEN

BACKGROUND: Tumor-associated antigens (TAAs) are known to be immunotherapy targets; thus tumor-sharing TAA may be used as a fusion hybrid partner to confer protection against subsequent tumor challenge. METHODS: The squamous cell carcinomas (SCCs), SCCVII and B4B8, were used in C3H/HEN mice: SCCVII (H-2(k)) is syngeneic, B4B8 (H-2(d)) is allogeneic. Experiments using tumor alone included hyperimmunization schedule, subdermal and intranodal routes. Mice were challenged 2 weeks later. Fusion hybrids were created from both SCC tumor cell lines and syngeneic dendritic cells (DCs). These were delivered intranodally for immunization, and mice were challenged with tumor 2 weeks later. RESULTS: Only syngeneic tumor given subdermally was able to protect after tumor challenge 2 weeks later. Hyperimmunization schedule did not alter these findings. However, fusion hybrid immunization from both allogeneic and syngeneic SCCs conferred protection after tumor challenge. CONCLUSIONS: Allogeneic tumor-DC fusion hybrids targeting TAA can protect against subsequent tumor challenge.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Células Dendríticas/trasplante , Células Híbridas/trasplante , Inmunoterapia/métodos , Animales , Carcinoma de Células Escamosas/inmunología , Fusión Celular , Línea Celular Tumoral , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Electroforesis , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Híbridas/inmunología , Interleucina-18/inmunología , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Distribución Aleatoria , Sensibilidad y Especificidad , Trasplante Homólogo
5.
Head Neck ; 32(6): 700-7, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19908319

RESUMEN

BACKGROUND: The aim of the present study was to evaluate the therapeutic efficacy of dendritic cell (DC)-tumor fusion hybrids with Toll-like receptor (TLR) agonists. METHODS: DC-tumor fusion hybrids were generated by electrofusion and injected into the inguinal lymph nodes of C57BL/6 mice with 3-day established pulmonary metastases. Paired TLR agonists polyinosine:polycytadilic acid [poly(I:C)] and cytosine-phosphate-guanine (CpG) were then injected intraperitoneally. Enzyme-linked immunosorbent assay (ELISA) was used to evaluate interleukin (IL)-12 production from the DC-tumor fusion hybrids in vitro. RESULTS: Fusion + TLR agonists (60 metastases) had significantly fewer metastases than did the untreated control (262 metastases, p = .0001) and fusion alone (150 metastases, p = .02). ELISA showed that the DC-tumor fusion hybrids yielded 90 pg of IL-12 after TLR stimulation compared with 1610 pg from dendritic cells alone. CONCLUSIONS: CpG and poly(I:C) administered as a third signal with fusion hybrids as described significantly reduce melanoma metastasis compared with fusion hybrids alone. Fusion hybrids do not appear to be a significant source for IL-12 secretion.


Asunto(s)
Fusión Celular/métodos , Células Dendríticas/inmunología , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Melanoma Experimental/terapia , Receptores Toll-Like/agonistas , Adyuvantes Inmunológicos , Animales , Estimulación Eléctrica , Ensayo de Inmunoadsorción Enzimática , Femenino , Células Híbridas/inmunología , Interleucina-12/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/secundario , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes de Fusión/inmunología , Organismos Libres de Patógenos Específicos , Vacunas Sintéticas
6.
J Immunol ; 181(5): 3291-300, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18714001

RESUMEN

Suppression of tumor-specific T cell sensitization is a predominant mechanism of tumor escape. To identify tumor-induced suppressor cells, we transferred spleen cells from mice bearing progressive MCA205 sarcoma into sublethally irradiated mice. These mice were then inoculated subdermally with tumor cells to stimulate T cell response in the tumor-draining lymph-node (TDLN). Tumor progression induced splenomegaly with a dramatic increase (22.1%) in CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSC) compared with 2.6% of that in normal mice. Analyses of therapeutic effects by the adoptive immunotherapy revealed that the transfer of spleen cells from tumor-bearing mice severely inhibited the generation of tumor-immune T cells in the TDLN. We further identified MDSC to be the dominant suppressor cells. However, cells of identical phenotype from normal spleens lacked the suppressive effects. The suppression was independent of CD4(+)CD25(+) regulatory T cells. Intracellular IFN-gamma staining revealed that the transfer of MDSC resulted in a decrease in numbers of tumor-specific CD4(+) and CD8(+) T cells. Transfer of MDSC from MCA207 tumor-bearing mice also suppressed the MCA205 immune response indicating a lack of immunologic specificity. Further analyses demonstrated that MDSC inhibited T cell activation that was triggered either by anti-CD3 mAb or by tumor cells. However, MDSC did not suppress the function of immune T cells in vivo at the effector phase. Our data provide the first evidence that the systemic transfer of MDSC inhibited and interfered with the sensitization of tumor-specific T cell responses in the TDLN.


Asunto(s)
Fibrosarcoma/inmunología , Ganglios Linfáticos/patología , Células Mieloides/patología , Escape del Tumor , Traslado Adoptivo , Animales , Antígeno CD11b , Línea Celular Tumoral , Femenino , Fibrosarcoma/patología , Fibrosarcoma/terapia , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Ratones , Células Mieloides/trasplante , Trasplante de Neoplasias , Receptores de Quimiocina , Linfocitos T/inmunología
7.
Cancer Res ; 68(11): 4045-9, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18519662

RESUMEN

Minimal requirements for generating effective immunity include the delivery of antigenic (signal 1) and costimulatory (signal 2) signals to T lymphocytes. Recently, a class of third signals, often delivered by antigen-presenting dendritic cells, has been shown to greatly enhance immune responses, especially against tumors. Among signal 3 factors, interleukin (IL)-12 is particularly effective and can be conditionally induced by agonists of Toll-like transmembrane receptors (TLR). In this study, we assessed the therapeutic effect of adjuvant TLR agonist administration upon the capacity of dendritic cell (DC)-tumor electrofusion hybrids to eradicate established MCA205 sarcomas in syngeneic mice. Paired, but not solitary combinations of polyinosine:polycytadilic acid (P[I:C]; TLR3 agonist) and CpG DNA (ODN1826l; TLR9 agonist) stimulated IL-12 secretion from DCs in vitro and synergized with vaccination to achieve potent tumor rejection. Therapeutic effects, however, required coadministration of paired TLR agonists and DC-tumor fusion hybrids. The administration of TLR agonists alone or with fusion vaccine induced transient splenomegaly but without apparent toxicity. The therapeutic effects of this immunization regimen were significantly abrogated through the neutralization of IL-12p70, indicating that production of this third signal was essential to the observed tumor regression. These results show the profound functional consequences of TLR cooperativity and further highlight the critical role of IL-12 in antitumor immunity.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Interleucina-12/biosíntesis , Receptores Toll-Like/agonistas , Vacunas/inmunología , Animales , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Femenino , Ratones , Ratones Endogámicos C57BL , Vacunas/administración & dosificación
8.
Blood ; 112(5): 1832-43, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18577706

RESUMEN

The clinical outcomes of dendritic cell (DC)-based immunotherapy remain disappointing, with DCs often displaying a tenuous capacity to complete maturation and DC1 polarization in the tumor host. Surprisingly, we observed that the capacity for successful DC1 polarization, including robust IL12p70 production, could be regulated by STAT-dependent events even prior to DC differentiation. Exposure of CD34(pos) cells to single-agent granulocyte-macrophage colony-stimulating factor (GMCSF) induced multilineage, STAT5-dependent differentiation, including DCs that failed to mature in the absence of further exogenous signals. In contrast, Flt3L induced nearly global differentiation of CD34(pos) cells into spontaneously maturing DCs. IL-6 synergized with Flt3L to produce explosive, STAT3-dependent proliferation of phenotypically undifferentiated cells that nevertheless functioned as committed DC1 precursors. Such precursors not only resisted many tumor-associated immunosuppressants, but also responded to tumor contact or TGFbeta with facilitated DC maturation and IL12p70 production, and displayed a superior capacity to reverse tumor-induced T-cell tolerance. GMCSF preempted Flt3L or Flt3L plus IL-6 licensing by blocking STAT3 activation and promoting STAT5-dependent differentiation. Paradoxically, following overt DC differentiation, STAT5 enhanced whereas STAT3 inhibited DC1 polarization. Therefore, nonoverlapping, sequential activation of STAT3 and STAT5, achievable by sequenced exposure to Flt3L plus IL-6, then GMCSF, selects for multilog expansion, programming, and DC1 polarization of tumor-competent DCs from CD34(pos) cells.


Asunto(s)
Antígenos CD34/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Diferenciación Celular/fisiología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Inmunoterapia Adoptiva , Interleucina-6/farmacología , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Proteínas Recombinantes , Factor de Transcripción STAT3/deficiencia , Factor de Transcripción STAT3/genética
9.
Arch Otolaryngol Head Neck Surg ; 134(6): 608-13, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18559727

RESUMEN

OBJECTIVE: To investigate the therapeutic efficacy of fused dendritic-tumor cell hybrids against murine squamous cell carcinoma (SCC). DESIGN: Squamous cell carcinoma VII is a poorly immunogenic murine SCC tumor in C3H/HEN (H-2(K)) mice. Subdermal tumors were established by inoculation in the mid abdomen of mice. Tumor diameters were measured with a Vernier caliper and used as an indication of treatment efficacy. Survival studies were performed on mice with 3-day pulmonary metastasis or subdermal tumors. Dendritic cells were generated from bone marrow and cultured for 8 days. Dendritic cells were harvested and mixed with cultured tumor cells in a 1:1 ratio. Cell fusion was achieved by exposing the cell mixture to an alternate electrical current to bring cells into alignment and close together, followed by a short direct electrical current pulse. SUBJECTS: Female C3H/HEN mice aged 8 to 12 weeks. INTERVENTIONS: Mice with 3-day established SCCVII tumors were vaccinated by inguinal intranodal injection of fusion cells (0.3 x 10(6) per side). To support the development of antitumor immunity, mice were given adjuvant injections intraperitoneally. Anti-OX40R monoclonal antibodies or interleukin 12 were used. Treatment groups included no treatment, anti-OX40R monoclonal antibodies or adjuvant IL-12 alone, fusion cells alone, and fusion cells with adjuvant treatment. MAIN OUTCOME MEASURES: Tumor size and overall survival. RESULTS: Mice treated with adjuvant treatment or fusion cells alone did not show a statistical difference in tumor growth when compared with controls. In contrast, fusion cells with adjuvant treatment demonstrated a significant decrease in tumor size when compared with nontreated mice (P < .001). Treatment with fusion cells also resulted in increased survival in the pulmonary metastasis and subdermal tumor models. CONCLUSION: Immunotherapy with fused dendritic-tumor cell hybrids can significantly affect 3-day established sSCC VII tumor growth.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Células Dendríticas/inmunología , Inmunoterapia , Células Tumorales Cultivadas/inmunología , Animales , Fusión Celular , Modelos Animales de Enfermedad , Femenino , Células Híbridas , Ratones , Ratones Endogámicos C3H
10.
J Immunol ; 180(4): 2140-8, 2008 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-18250420

RESUMEN

Engagement of OX40 greatly improves CD4 T cell function and survival. Previously, we showed that both OX40 engagement and CTLA-4 blockade led to enhanced CD4 T cell expansion, but only OX40 signaling increased survival. To identify pathways associated with OX40-mediated survival, the gene expression of Ag-activated CD4 T cells isolated from mice treated with anti-OX40 and -CTLA-4 was compared. This comparison revealed a potential role for IL-12 through increased expression of the IL-12R-signaling subunit (IL-12Rbeta2) on T cells activated 3 days previously with Ag and anti-OX40. The temporal expression of IL-12Rbeta2 on OX40-stimulated CD4 T cells was tightly regulated and peaked approximately 4-6 days after initial activation/expansion, but before the beginning of T cell contraction. IL-12 signaling, during this window of IL-12Rbeta2 expression, was required for enhanced T cell survival and survival was associated with STAT4-specific signaling. The findings from these observations were exploited in several different mouse tumor models where we found that the combination of anti-OX40 and IL-12 showed synergistic therapeutic efficacy. These results may lead to the elucidation of the molecular pathways involved with CD4 T cell survival that contribute to improved memory, and understanding of these pathways could lead to greater efficacy of immune stimulatory Abs in tumor-bearing individuals.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Interleucina-12/fisiología , Receptores OX40/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/inmunología , Femenino , Interleucina-12/deficiencia , Interleucina-12/genética , Subunidad p35 de la Interleucina-12/deficiencia , Subunidad p35 de la Interleucina-12/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neoplasias de la Próstata/inmunología , Receptores de Interleucina-12/biosíntesis , Receptores de Interleucina-12/deficiencia , Receptores de Interleucina-12/genética , Receptores OX40/agonistas , Receptores OX40/metabolismo , Sarcoma Experimental/inmunología , Transducción de Señal/inmunología
11.
Clin Immunol ; 127(1): 66-77, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18262845

RESUMEN

Curative immunotherapy against spontaneous metastases of poorly immunogenic tumors has been difficult to demonstrate, but it is highly relevant to clinical disease conditions. The 4T1 mammary carcinoma shares many characteristics of human mammary cancer. Here, mice with 4T1 spontaneous metastases were treated effectively with a combination of dendritic (DC)-tumor hybrid vaccination and adoptive transfer of tumor-draining lymph node-derived immune T cells. This strategy significantly prolonged survival and cured some mice. In this model, the combined immunotherapy induced a dramatic increase of T cells in the lung where metastases were located and in the spleen where tumor was not present. The mechanism of increasing numbers of T cells is likely attributed to the ability of DC-tumor hybrids to stimulate vigorous proliferation of adoptively transferred T cells rather than to promote their infiltration into tumor-harboring and lymphoid organs. Taken together, the combined approach may be useful for clinical development of cancer immunotherapy.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Neoplasias Mamarias Experimentales/terapia , Metástasis de la Neoplasia/terapia , Linfocitos T/inmunología , Animales , Fusión Celular , Células Dendríticas/trasplante , Femenino , Células Híbridas/inmunología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Células Tumorales Cultivadas
12.
Am J Otolaryngol ; 28(6): 375-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17980767

RESUMEN

PURPOSE: Laryngeal transplantation has not been widely accepted because of concerns regarding accelerated tumor recurrences in the setting of nonspecific immunosuppression. Allotransplantation could potentially be offered to patients if immunosuppressive therapy could be demonstrated to exert tumor suppressive properties. Preliminary reports have demonstrated an antiproliferative effect of everolimus (RAD), a derivative of the immunosuppressant rapamycin. MATERIALS AND METHODS: Forty-five 10-week-old inbred C57BL/6N (B6) mice were injected subcutaneously with 1 x 10(6) MCA205 sarcoma cells. On the third postinoculation day, the mice were divided into 4 treatment groups, undergoing daily gavage with RAD at 0, 0.2, 1.0, and 5.0 mg/kg per day for 10 consecutive days. Thereafter, treatment with RAD was discontinued and tumor size was measured every 2 days during treatment and biweekly until sacrifice on the 31st postinoculation day. Whole-blood trough levels (C(min)) were measured for each group. RESULTS: Mean tumor diameter among the control animals and the mice treated with RAD 0.2 mg/kg per day demonstrated no significant difference (P > .07). Groups treated with RAD 1 and 5 mg/kg per day demonstrated significant growth inhibition between the 7th and the 23rd postinoculation days (P < .0001), with no significant differences being noted between these two groups (P > .09). Mean tumor suppressive whole-blood C(min)'s for the 1 and 5 mg/kg per day groups were 75.6 and 368.9 pg/microL, respectively. CONCLUSIONS: RAD delivered at immunosuppressive doses of 1 and 5 mg/kg per day resulted in significant growth restriction of a fibrosarcoma in a murine model.


Asunto(s)
Fibrosarcoma/tratamiento farmacológico , Fibrosarcoma/patología , Inmunosupresores/administración & dosificación , Sirolimus/análogos & derivados , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Neoplasias de los Tejidos Blandos/patología , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Everolimus , Ratones , Ratones Endogámicos C57BL , Sirolimus/administración & dosificación , Carga Tumoral
14.
Blood ; 109(11): 4865-76, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17284532

RESUMEN

The importance of CD4+ Th1 cells during the effector phase of the antitumor response has been overshadowed by emphasis on CD8+ cytotoxic T lymphocytes (CTLs). To determine their respective functions, we purified antigen-primed T cells from tumor-draining lymph nodes and separately activated CD4+ and CD8+ subsets in vitro. Adoptive transfer of CD4+ T effector cells (T(E)s) combined with CD8+ T(E)s provided synergistic therapy for mice bearing subcutaneous, intracranial, or advanced pulmonary metastases. CD4+ T(E)s augmented IFN-gamma production by CD8+ T(E)s when cells were stimulated by tumor digest-containing antigen-presenting cells (APCs). CD4+ T(E)s infiltrated and proliferated extensively in pulmonary tumors, while also stimulating tumor antigen-specific CD8+ T cells. By contrast, CD8+ T(E)s showed minimal intratumoral proliferation in the absence of CD4+ cells or when systemically transferred CD4+ cells were prevented from infiltrating pulmonary tumors by pretreatment with pertussis toxin. Irradiation of CD4+ T cells immediately prior to adoptive transfer abrogated their intratumoral proliferation and direct antitumor efficacy but did not block their capacity to stimulate intratumoral CD8+ T(E) proliferation or tumor regression. These results highlight the importance of cross-presentation of tumor antigens during the effector phase of immunotherapy and suggest that approaches to stimulate CD4+ T(E) function and boost APC cross-presentation within tumors will augment cancer immunotherapy.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos , Neoplasias Pulmonares/metabolismo , Linfocitos T Citotóxicos/inmunología , Animales , Proliferación Celular , Separación Celular , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia
15.
J Immunol ; 178(4): 2278-86, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17277133

RESUMEN

The role of tumor-produced chemokines in the growth of malignancies remains poorly understood. We retrieved an in vivo growing MCA205 fibrosarcoma and isolated tumor cell clones that produce both CXCL9/monokine induced by IFN-gamma (Mig) and CXCL10/IFN-gamma-inducible protein 10 following stimulation with IFN-gamma and clones that produce IFN-gamma-inducible protein 10 but not Mig. The Mig-deficient variants grew more aggressively as cutaneous tumors in wild-type mice than the Mig-producing tumor cells. The growth of Mig-expressing, but not Mig-deficient, tumor cells was suppressed by NK and T cell activity. Transduction of Mig-negative variants to generate constitutive tumor cell production of Mig resulted in T cell-dependent rejection of the tumors and in induction of protective tumor-specific CD8(+) T cell responses to Mig-deficient tumors. The results indicate a critical role for tumor-derived Mig in T cell-mediated responses to cutaneous fibrosarcomas and suggest the loss of Mig expression as a mechanism used by tumor cells to evade these responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiocinas CXC/inmunología , Fibrosarcoma/inmunología , Regulación Neoplásica de la Expresión Génica/inmunología , Interferón gamma/inmunología , Monocinas/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias Cutáneas/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Quimiocina CXCL9 , Quimiocinas CXC/biosíntesis , Quimiocinas CXC/deficiencia , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Noqueados , Monocinas/biosíntesis , Monocinas/deficiencia , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/deficiencia , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Escape del Tumor/genética , Escape del Tumor/inmunología
16.
Crit Rev Immunol ; 27(5): 463-83, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18197808

RESUMEN

Cancer immunotherapy using fusion hybrid cells generated from dendritic cells (DCs) and tumor cells may be more effective than other DC-based vaccines. DC-tumor fusion potentially confers not only the DCs' antigen-presenting functionality but also a continuing source of endogenous tumor antigens for major-histocompatibility-complex-restricted T-cell sensitization. In animal models, many investigators demonstrated that vaccination with fusion hybrids was protective against tumor challenge and therapeutic, resulting in the regression of established tumors. In clinical trials for patients with a variety of metastatic diseases, fusion hybrid vaccines were well tolerated, but the overall objective response rate was only 10.9%. Careful scrutiny of a large number of publications revealed that, in most cases, no definitive evidence of heterokaryonic fusion cell formation was found. Further corroboration of this conclusion comes from reports that fusion hybrids generated from autologous (syngeneic) and allogeneic DCs displayed equivalent immunological function and therapeutic effects in vitro and in vivo. This puzzling finding suggests that effective fusion immunotherapy depends on tumor antigen scavenging and presentation by antigen-presenting cells (APCs) of host origin and is in violation of the basic tenet of the principle of DC function. We believe that conclusions drawn from reported clinical trials have not properly evaluated the efficacy of the DC-tumor hybrid vaccine, and therefore, they neither confirm nor disclaim the potential benefits that may be derived from this form of immunotherapy.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Fusión Celular/métodos , Humanos , Células Híbridas/inmunología , Inmunoterapia
17.
Cancer Metastasis Rev ; 25(2): 233-42, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16770535

RESUMEN

Regional lymph nodes are the first site for melanoma metastases. The sentinel node (SN), on the direct lymphatic drainage pathway, which usually harbors first metastases, demonstrates significant suppression in its ability to respond to antigenic stimulation. This down-regulation of SN immunity is likely the basis of its susceptibility to tumor metastases, suggesting a potential role of the immune system in the control of malignant tumors. Despite immune dysfunction in the SN, phase II trials of systemic post-operative immunotherapy with a polyvalent melanoma vaccine developed at the John Wayne Cancer Institute showed improved 5-year overall survival in patients with melanoma metastatic to regional nodes. However, most immunotherapy clinical trials have failed to demonstrate a significant clinical response, and analyses of immune responses to tumor-associated antigens that correlate clinical responses have not been established. Therefore, refinements in assay methodologies and improvements in vaccine designs are critical to the success of cancer immunotherapy. Antigen presentation by dendritic cells (DCs) is the most potent means to initiate a T cell immunity. Dendritic cell-based immunotherapies have been vigorously attempted in the past decade. To improve the immunogenicity of cancer vaccines, we recently generated heterokaryons of DCs and tumor cells by electrofusion. The fusion hybrids retained their full antigen-presenting capacity and all natural tumor antigens. In pre-clinical animal experiments, a single injection of the DC-tumor fusion hybrids was sufficient to mediate the regression of tumors established in the lung, skin and brain. Most interestingly, successful therapy required the delivery of fusion hybrids directly into lymphoid organs such as lymph nodes. A clinical trial is now being carried out to test the immunogenicity and therapeutic effects of fusion hybrids for the treatment of metastatic melanoma.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Melanoma/inmunología , Melanoma/terapia , Neoplasias/inmunología , Neoplasias/terapia , Animales , Fusión Celular , Ensayos Clínicos como Asunto , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Humanos , Tolerancia Inmunológica , Inmunoterapia , Ganglios Linfáticos , Metástasis Linfática , Melanoma/secundario , Sobrevida
18.
Laryngoscope ; 116(5): 814-20, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16652094

RESUMEN

OBJECTIVE: Everolimus (RAD) is an mTOR inhibitor closely related to rapamycin. A potent immunosuppressive agent, it has also shown evidence of antineoplastic properties. SCC VII is a spontaneously arising murine squamous cell carcinoma line. This study examines the effect of everolimus on SCC VII proliferation. The data may provide support for the use of everolimus in transplant recipients with a history of malignancy. METHODS: A dose efficacy study was conducted that used a murine model of intradermal tumor growth and pulmonary metastases. The development of intradermal tumors and pulmonary metastases were studied. Of 80 total mice, 40 received intradermal injection of 1 x 10 SCC VII cells and 40 received intravenous injection of 1 x 10 cells to establish pulmonary metastases. Within each group, animals were subdivided into four subgroups that received 1) 1 mg/kg everolimus twice a day, 2) 0.5 mg/kg everolimus twice a day, 3) 7.5 mg/kg cyclosporine per day, and 4) no treatment. Intradermal tumors were measured three times per week. Animals receiving an intravenous tumor injection were killed after 17 days and pulmonary metastases were quantified. Medication trough levels were measured in all treated animals. RESULTS: Everolimus showed statistically significant tumor inhibition at 1.0 mg/kg twice a day and 0.5 mg/kg twice a day when compared with animals treated with cyclosporine and with untreated animals (P < .0001). Tumor inhibition was evident in both models studied (intradermal tumors and pulmonary metastasis generation). CONCLUSIONS: Everolimus provides potent tumor inhibition in animals inoculated with SCC VII cells. Inhibition of both local and distant spread of disease is evident. Although most immunosuppressives are known to potentiate neoplastic disease, this study supports the use of everolimus immunosuppression in the face of prior malignancy. This data has significant implication for laryngeal transplantation after laryngectomy.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/secundario , Neoplasias Pulmonares/secundario , Sirolimus/análogos & derivados , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Animales , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Everolimus , Femenino , Inmunosupresores/farmacología , Inyecciones Intravenosas , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C3H , Trasplante de Neoplasias , Probabilidad , Distribución Aleatoria , Valores de Referencia , Sensibilidad y Especificidad , Sirolimus/farmacología , Células Tumorales Cultivadas
19.
Cell Immunol ; 243(1): 30-40, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17207783

RESUMEN

Dendritic-tumor heterokaryons generated by electrofusion are highly immunogenic. In animal studies, a single vaccination was therapeutic for tumors established in the lung, skin, and brain. However, effective therapy required a third signal which could be provided by exogenous IL-12 or the agonistic anti-OX-40R monoclonal antibody (mAb). In this study, we investigated the mechanism and mode of actions of these two seemingly distinct adjuvants. In immunotherapy of the MCA205 sarcoma, administration of the neutralizing anti-IL-12 mAb nearly completely blocked the adjuvant effect of IL-12, but had minimal inhibitory effects on anti-OX-40R mAb. By contrast, in vivo administration of the antagonistic anti-OX-40L mAb inhibited the adjuvant effects of both IL-12 and anti-OX-40R mAb. Thus, a common pathway of endogenous OX-40 interaction is critical for the development of a therapeutic immune response. Analysis of the third signal mechanism revealed that in the absence of an adjuvant, vaccination with fusion hybrids led to IL-10 production without eliciting IFN-gamma secreting cells. The addition of IL-12 to vaccination suppressed IL-10 production and initiated sensitization of specific IFN-gamma secreting cells, resulting in a type 1-like antitumor immunity. These findings underscore the significance of the third signal in the design of dendritic cell-based cancer vaccines.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/inmunología , Inmunidad Celular , Interleucina-12/farmacología , Ligando OX40/farmacología , Sarcoma Experimental/terapia , Animales , Anticuerpos Monoclonales/inmunología , Fusión Celular , Células Dendríticas/metabolismo , Femenino , Interferón gamma/metabolismo , Interleucina-10/metabolismo , Interleucina-12/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ligando OX40/uso terapéutico , Vacunación/métodos
20.
Cancer Res ; 65(20): 9547-54, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16230420

RESUMEN

T-cell adoptive immunotherapy for stringent murine tumor models, such as intracranial, s.c., or advanced pulmonary metastases, routinely uses lymphodepletive conditioning regimens before T-cell transfer, like recent clinical protocols. In this study, we examined whether host lymphodepletion is an obligatory component of curative T-cell therapy; we also examined the mechanism by which it augments therapy. Mice bearing intracranial, s.c., or 10-day pulmonary metastases of MCA 205 received total body irradiation conditioning or were nonirradiated before i.v. transfer of tumor-reactive T cells. Total body irradiation was not required for immunologically specific curative therapy and induction of memory provided that a 3- to 12-fold higher T-cell dose was administered. The mechanism involved enhanced intratumoral proliferation of T-effector cells in total body irradiation-conditioned recipients. In this tumor model, intratumoral T(reg) cells were not detected; consequently, intratumoral T-effector cells produced identical amounts of IFN-gamma upon ex vivo antigen stimulation irrespective of total body irradiation conditioning. Thus, host lymphodepletion augments T-cell immunotherapy through enhanced antigen-driven proliferation of T-effector cells, but curative therapy can be achieved in nonconditioned hosts by escalation of T-cell dose. These data provide a rationale for dose escalation of T-effector cells in situations where single or repeated lymphodepletion regimens are contraindicated.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Linfocitos T/inmunología , Linfocitos T/efectos de la radiación , Irradiación Corporal Total , Animales , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Complejo CD3/inmunología , Terapia Combinada , Epítopos de Linfocito T/inmunología , Femenino , Fibrosarcoma/inmunología , Fibrosarcoma/secundario , Fibrosarcoma/terapia , Interleucina-2/inmunología , Interleucina-7/inmunología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA