Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Br J Ophthalmol ; 104(4): 524-528, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31371315

RESUMO

BACKGROUND/AIMS: Survival after diagnosis of metastasis from uveal melanoma is poor. Identifying individuals at high risk for metastasis and developing adjuvant therapy to prevent clinically apparent metastasis could improve survival. We conducted an adjuvant trial of sequential, low-dose dacarbazine (DTIC) and interferon-alpha-2b (IFN-α-2b) in patients with cytogenetic high-risk uveal melanoma. METHODS: Patients diagnosed with iris, ciliary body or choroidal melanoma with high-risk tumour cytogenetics (monosomy 3) were offered adjuvant treatment with low-dose DTIC and IFN-α-2b following primary therapy. Eligible but not enrolled patients were observed for comparison. DTIC was administered at 850 mg/m2 intravenously on days 1 and 28. IFN-α-2b was administered at 3 million units three times a week subcutaneously for 24 weeks beginning at week 9. Hepatic imaging was performed prior to adjuvant therapy and then at least every 6 months. Survival data were collected for 5 years after enrolment. RESULTS: 33 patients (22%) were enrolled (treatment group), 29 (19%) were eligible but did not enrol (observation group) and 88 (59%) were not eligible. The 5-year metastasis-free survival (MFS) was 64%±9% for treated and 33%±10% for observed patients (p=0.05). The 5-year overall survival (OS) rate was 66%±9% for treated and 37%±10% for observed patients (p=0.02). CONCLUSIONS: When adjusted for differences in age, tumour size and initial treatment, survival between treated and observed patients was no longer significant (p=0.56 MFS and p=0.92 OS). Differences in baseline tumour characteristics between treated and observed patients can influence interpretation of results. TRIAL REGISTRATION NUMBER: NCT01100528.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Dacarbazina/administração & dosagem , Interferon-alfa/administração & dosagem , Melanoma/tratamento farmacológico , Melanoma/mortalidade , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/mortalidade , Adulto , Idoso , Idoso de 80 Anos ou mais , Quimioterapia Adjuvante , Intervalo Livre de Doença , Feminino , Humanos , Infusões Intravenosas , Injeções Subcutâneas , Interferon alfa-2 , Masculino , Melanoma/patologia , Pessoa de Meia-Idade , Estudos Prospectivos , Taxa de Sobrevida , Neoplasias Uveais/patologia , Adulto Jovem
2.
Clin Exp Metastasis ; 33(8): 829-838, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27565163

RESUMO

Natural killer (NK) cells are implicated in the control of metastasis in uveal melanoma, a process that has been ascribed to its cancer stem cell subpopulation. NK cell activation is regulated by specific microRNA (miR). The NK cell sensitivity and regulatory miR production of uveal melanoma cancer stem cells was examined. Cancer stem cells enriched from aggressively metastatic MUM2B uveal melanoma cells by selecting CD271+ cells or propagating as non-adherent spheres in stem-cell supportive were more resistant to NK cell cytolysis than cancer stem cells enriched from less aggressively metastatic OCM1 uveal melanoma cells. Both MUM2B and OCM1 cells expressed and secreted NK cell regulatory miRs, including miR 146a, 181a, 20a, and 223. MUM2B cells expressed and secreted miR-155; OCM1 cells did not. Transfecting MUM2B cells with anti-miR-155 increased NK cell sensitivity. CD271+ cells were identified in the blood of patients with metastatic uveal melanoma and were characterized by low expression of melanocyte differentiation determinants and by the ability to form non-adherent spheres in stem-cell supportive media. These cells also expressed NK cell regulatory miRs, including miR-155. These results indicate that uveal melanoma cancer stem cells can vary in their sensitivity to NK cell lysis and their expression of NK cell regulatory miRs. Circulating CD271+ cells from patients with metastatic uveal melanoma manifest cancer stem cell features and express miRs associated with NK cell suppression, including miR-155, that may contribute to metastatic progression.


Assuntos
Células Matadoras Naturais/patologia , Melanoma/genética , MicroRNAs/genética , Células-Tronco Neoplásicas/metabolismo , Neoplasias Uveais/genética , Adapaleno/imunologia , Adapaleno/metabolismo , Citotoxicidade Imunológica/imunologia , Regulação Neoplásica da Expressão Gênica , Humanos , Células Matadoras Naturais/metabolismo , Melanócitos/patologia , Melanoma/patologia , MicroRNAs/biossíntese , Células-Tronco Neoplásicas/patologia , Transfecção , Neoplasias Uveais/patologia
3.
Clin Epigenetics ; 8: 80, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27453764

RESUMO

BACKGROUND: Epigenetic events mediated by methylation and histone modifications have been associated with the development of metastasis in patients with uveal melanoma. The role of epigenetic events mediated by microRNA (miR) is less clear. Tumor and plasma miR expression was examined in patients with primary uveal melanoma with tumor monosomy-3, a predictor of metastasis. RESULTS: miR profiling of tumors by microarray found six miRs over-expressed and 19 under-expressed in 33 tumors with monosomy-3 compared to 22 without. None of the miRs differentially expressed in tumors with and without monosomy-3 was differentially expressed in tumors with and without tumor infiltrating lymphocytes. Tumors manifesting monosomy-3 were also characterized by higher levels of TARBP2 and DDX17 and by lower levels of XPO5 and HIWI, miR biogenesis factors. miR profiling of plasma by a quantitative nuclease protection assay found elevated levels of 11 miRs and reduction in four in patients with tumor monosomy-3. Only three miRs differentially expressed in the tumor arrays were detectable in plasma. miRs implicated in uveal melanoma development were not differentially expressed. Elevated plasma levels in patients with tumor monosomy-3 of miR-92b, identified in the tumor array, and of miR-199-5p and miR-223, identified in the plasma array, were confirmed by quantitative real-time polymerase chain reaction. Levels were also higher in patients compared to normal controls. CONCLUSIONS: These results support a role for epigenetic mechanisms in the development of metastasis in patients with uveal melanoma and the analysis of miRs as biomarkers of metastatic risk. They also suggest that potentially useful blood miRs may be derived from the host response as well as the tumor.


Assuntos
Cromossomos Humanos Par 3/genética , Melanoma/genética , MicroRNAs/genética , Monossomia , Neoplasias Uveais/genética , Epigênese Genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma/sangue , MicroRNAs/sangue , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Neoplasias Uveais/sangue
4.
Melanoma Res ; 25(6): 510-8, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26397048

RESUMO

Antigens that may be involved in the immune response to uveal melanoma have not been identified. Cellular and humoral responses to melanoma differentiation antigens, as well as to BRCA1-associated protein 1 (BAP1) and α-enolase, alterations of which are associated with metastatic disease, were examined in patients with uveal melanoma. Blood was collected from 66 patients with primary and 13 patients with metastatic uveal melanoma. These included 11 patients treated with immunotherapy. Peripheral blood mononuclear cells were stimulated with gp100, MART-1, tyrosinase, NY-ESO-1, BAP1, and α-enolase peptides and/or proteins, and cytokine production was assessed by bead array or enzyme-linked immunosorbent assay. Autoantibodies to the protein were assessed by enzyme-linked immunosorbent assay. A cellular or humoral response to one or more of the antigens was observed in 23% of the primary and 62% of the metastatic patients tested. Th1 and Th2 cellular and humoral responses to gp100, MART-1, and tyrosinase were observed in primary and metastatic patients. Cellular responses to NY-ESO-1 were not observed nor were Th17-associated responses. Cellular and humoral responses to BAP1 and α-enolase were also observed, predominantly in primary patients with tumor monosomy-3 and in metastatic patients. Individual patients treated with immunotherapy developed new reactivity to MART-1, tyrosinase, and/or α-enolase. Patients with primary and metastatic uveal melanomas manifest spontaneous immune responses to melanoma differentiation antigens, BAP1, and α-enolase. Both Th1-associated and Th2-associated responses are observed and can be modified by therapy. These results may help the development and monitoring of immunotherapy and studies of immune surveillance in uveal melanoma.


Assuntos
Antígenos de Neoplasias/imunologia , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Melanoma/imunologia , Neoplasias Uveais/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Ensaio de Imunoadsorção Enzimática , Humanos
5.
Mol Immunol ; 58(2): 182-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24370793

RESUMO

BACKGROUND: The immune response has been implicated in the control of uveal melanoma progression. Epigenetic mechanisms mediated by specific microRNAs (miRs) regulate immune responses. METHODS: Blood was drawn from six patients with uveal melanoma followed from diagnosis, at which time there was no clinical or radiographic evidence of metastasis, until metastasis manifested. Circulating T cell, natural killer (NK), natural killer T (NKT), and myeloid suppressor cell populations were assessed by flow cytometry. CD3(+), CD15(+), and CD56(+) cells were isolated using immunomagnetic beads. Plasma and cellular levels of immune regulatory miRs were determined by quantitative polymerase chain reaction assays. RESULTS: The development of metastasis was associated with decreases in circulating CD3(-)CD56(dim) NK cells and CD8(+) and double-negative CD3(+)CD56(+) NKT cells. ICOS(+)CD4(+)FoxP3(+) T regulatory cells and CD11b(+)CD14(-)CD15(+) myeloid suppressor cells increased. Plasma levels of miR-20a, 125b, 146a, 155, 181a, and 223 were higher in the study patients at diagnosis compared to controls. Plasma levels of miR-20a, 125b, 146a, 155, and 223 increased, and miR-181a decreased when metastasis manifested. Alterations in immune regulatory miRs were also observed in CD3(+), CD15(+), and CD56(+) cell populations. CONCLUSIONS: The development of metastasis in uveal melanoma is associated with changes in immune effector and regulatory cells consistent with lessening tumor immune surveillance. These changes are associated with changes in plasma and cellular levels of immune regulatory miRs. The results may help guide uveal melanoma immunotherapy and biomarker development.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Melanoma/sangue , Melanoma/patologia , MicroRNAs/sangue , Neoplasias Uveais/sangue , Neoplasias Uveais/patologia , Idoso , Estudos de Casos e Controles , Feminino , Humanos , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica
6.
Melanoma Res ; 23(1): 1-7, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23196330

RESUMO

Prognostic blood biomarkers for patients with uveal melanoma have not been identified. Tumor monosomy-3 is strongly associated with the development of metastatic disease. Tumor expression of human leukocyte antigen class I molecules and insulin-like growth factor (IGF)-1 receptor has also been associated with the development of metastatic disease. The relationship of blood levels of the human leukocyte antigen-class-I-associated ß-2 microglobulin (ß2M), IGF-1, and its binding protein, IGFBP-3, with tumor monosomy-3 was evaluated. Blood was drawn from patients with a clinical diagnosis of primary uveal melanoma without metastatic disease before fine-needle aspiration biopsy at the time of brachytherapy or enucleation. Tumor chromosome 3 status was determined by fluorescence in-situ hybridization. ß2M, IGF-1, and IGFBP-3 levels were determined using enzyme-linked immunosorbent assays. A total of 76 patients were studied; 47 (62%) underwent brachytherapy and 29 (38%) underwent enucleation. Thirty-three (43%) of the tumors manifested monosomy-3. Most tumors were large, located in the choroid, mixed cell type, and nuclear grade 2. Most tumors did not manifest extraocular extension. Blood levels of IGF-1 and IGFBP-3 were not associated with tumor monosomy-3. In contrast, increases in blood ß2M (P≤0.02) were associated with tumor monosomy-3. The independent association of increased blood level of ß2M and tumor monosomy-3 status was confirmed in multivariable analysis. In conclusion, measurement of blood levels of ß2M in patients with primary uveal melanoma may have prognostic value and may help guide surveillance and adjuvant therapy recommendations.


Assuntos
Biomarcadores Tumorais/sangue , Deleção Cromossômica , Melanoma/genética , Melanoma/patologia , Neoplasias Uveais/genética , Neoplasias Uveais/patologia , Microglobulina beta-2/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/genética , Braquiterapia , Cromossomos Humanos Par 3 , Enucleação Ocular , Feminino , Humanos , Proteína 3 de Ligação a Fator de Crescimento Semelhante à Insulina/sangue , Fator de Crescimento Insulin-Like I/metabolismo , Modelos Logísticos , Masculino , Melanoma/terapia , Pessoa de Meia-Idade , Análise Multivariada , Gradação de Tumores , Prognóstico , Neoplasias Uveais/terapia , Adulto Jovem
7.
J Transl Med ; 10: 241, 2012 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-23217102

RESUMO

BACKGROUND: Blood biomarkers are needed to monitor anti-angiogenic treatments for cancer. The association of blood levels of microRNAs (miRs) implicated in angiogenesis with circulating endothelial cells (CEC) and with angiogenic proteins was examined in patients administered drugs with anti-angiogenic activity. METHODS: Blood was collected from patients with uveal melanoma enrolled on an adjuvant therapy trial in which they were treated sequentially with dacarbazine and interferon-alfa-2b. Plasma levels of nine angioregulatory miRs, miR-16, 20a, 106a, 125b, 126, 146a, 155, 199a, and 221, were determined by quantitative real time polymerase chain reaction; CEC, by semi-automated immunomagnetic; and plasma angiogenic proteins, by enzyme linked immunosorbent assays. RESULTS: Levels of miR-199a were positively correlated and miR-106a negatively correlated with CEC pre-therapy. Decreases in miR-126 and miR-199a and increases in miR-16 and miR-106a were observed after interferon-alfa-2b, but not after dacarbazine. CEC also increased after treatment with interferon but not after treatment with dacarbazine. Levels of miRs did not correlate with levels of vascular endothelial growth factor, basic fibroblast growth factor, and interleukin-8. Angiogenic proteins also did not change significantly with treatment. CONCLUSIONS: Blood levels of specific angioregulatory miRs are associated with CEC, and changes in specific angioregulatory miRs parallel increases in CEC after treatment with interferon-alfa-2b. Blood levels of specific angioregulatory miRs are not associated with levels of angiogenic proteins. miRs warrant further evaluation as blood biomarkers of angiogenesis.


Assuntos
Proteínas Angiogênicas/sangue , Movimento Celular , Dacarbazina/uso terapêutico , Células Endoteliais/patologia , Interferon-alfa/uso terapêutico , MicroRNAs/sangue , Movimento Celular/efeitos dos fármacos , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Células Endoteliais/efeitos dos fármacos , Humanos , Interferon alfa-2 , Interferon-alfa/administração & dosagem , Interferon-alfa/farmacologia , Melanoma/sangue , Melanoma/irrigação sanguínea , Melanoma/tratamento farmacológico , Melanoma/patologia , Neovascularização Patológica/sangue , Neovascularização Patológica/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Neoplasias Uveais/sangue , Neoplasias Uveais/irrigação sanguínea , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/patologia
8.
J Immunother ; 35(7): 587-94, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22892455

RESUMO

Recombinant human (rh) granulocyte/macrophage colony stimulating factor (GM-CSF) has demonstrated antitumor immunologic activity in prostate and other cancers. Dosing has been empiric, and biomarkers of effect have not been established. Eight patients with biochemical relapse of prostate cancer were randomized into group A, in which they received rhGM-CSF at 250 µg/m days 1-14 of a 28-day cycle, and 8 were randomized into group B, in which they received 250 µg 3 times a week continuously. Blood dendritic cell (DC), immune suppressor cell, cytokine, and microRNA (miR) levels were examined using flow cytometric, enzyme-linked immunosorbent, and/or polymerase chain reaction-based assays. Group A had greater increases in myeloid DC and in granulocyte and monocytes. In croup B, plasmacytoid DC decreased. In group A, DC production of interleukin-12 relative to interleukin-10 decreased; this ratio increased in group B. Increases in myeloid-derived suppressor cells were observed in both the groups with increases greater in group A. Increases in regulatory T cells and in serum tumor necrosis and vascular endothelial growth factors were only observed in group A. Serum miR-155 decreased in group A. An increase in serum miR-223 and a decrease in miR-125b and miR-146a were observed in group B. The dosing of rhGM-CSF influences immune and miR effects. More DC activation and fewer myeloid-derived suppressor and regulatory T cells are observed when administered at lower doses intermittently and continuously compared with when administered at higher doses daily and cyclically. Serum levels of miRs are potentially useful biomarkers of these effects.


Assuntos
Adenocarcinoma/tratamento farmacológico , Células Dendríticas/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , MicroRNAs/efeitos dos fármacos , Células Mieloides/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Proteínas Recombinantes/administração & dosagem , Linfócitos T Reguladores/efeitos dos fármacos , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Idoso , Biomarcadores Farmacológicos/metabolismo , Protocolos Clínicos , Citocinas/metabolismo , Células Dendríticas/imunologia , Cálculos da Dosagem de Medicamento , Humanos , Terapia de Imunossupressão , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Células Mieloides/imunologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/imunologia , Linfócitos T Reguladores/imunologia
9.
Cancer Immunol Immunother ; 61(9): 1441-50, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22310929

RESUMO

BACKGROUND: Low doses of the demethylating agent decitabine have been shown to enhance the sensitivity of tumors to immune effector cells and molecules through upregulation of tumor antigen presentation and apoptotic pathways. Effects on host immune effector and suppressor responses have not been well characterized. METHODS: Mice bearing B16 melanoma were treated with low-dose decitabine, cytokine, interleukin-2 (IL-2), toll-like receptor 9 agonist ODN1826, and/or a viral vectored vaccine targeting the melanoma antigen Trp2. Lymphoid and myeloid effector and suppressor cells were examined both systemically and intratumorally with functional, flow cytometric, and polymerase chain reaction-based assays. RESULTS: Enhancement of tumor growth delay was observed when decitabine was applied sequentially but not concurrently with IL-2. In contrast, complete responses and prolonged survival were observed when decitabine was applied with ODN1826 as therapy and with ODN1826 as a Trp2 vaccine adjuvant. Decitabine decreased natural killer and antigen-specific cellular immune responses when administered concurrently with IL-2 and with ODN1826; the Th1-associated transcription factor Tbet also decreased. T regulatory cells were not affected. When applied concurrently with ODN1826, decitabine increased macrophage cytotoxicity, M1 polarization, and dendritic cell activation. Myeloid-derived suppressor cells were reduced. CONCLUSION: Low-dose decitabine promotes both anti- and pro-tumor host immune responses to immunotherapeutics in melanoma-bearing mice. Macrophage effector and dendritic cell activation increase, and myeloid suppressor cells decrease. Lymphoid effector responses, however, can be inhibited.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Azacitidina/análogos & derivados , Imunoterapia/métodos , Interleucina-2/farmacologia , Melanoma Experimental/imunologia , Melanoma Experimental/terapia , Oligodesoxirribonucleotídeos/farmacologia , Animais , Azacitidina/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada , Citocinas/biossíntese , Citocinas/imunologia , Decitabina , Relação Dose-Resposta a Droga , Feminino , Humanos , Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Melanoma Experimental/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos/imunologia , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/imunologia
10.
Int J Cancer ; 131(1): 18-29, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21796622

RESUMO

Apoptosis genes, such as TP53 and p16/CDKN2A, that mediate responses to cytotoxic chemotherapy, are frequently nonfunctional in melanoma. Differentiation may be an alternative to apoptosis for inducing melanoma cell cycle exit. Epigenetic mechanisms regulate differentiation, and DNA methylation alterations are associated with the abnormal differentiation of melanoma cells. The effects of the deoxycytidine analogue decitabine (5-aza-2'-deoxycytidine), which depletes DNA methyl transferase 1 (DNMT1), on melanoma differentiation were examined. Treatment of human and murine melanoma cells in vitro with concentrations of decitabine that did not cause apoptosis inhibited proliferation accompanied by cellular differentiation. A decrease in promoter methylation, and increase in expression of the melanocyte late-differentiation driver SOX9, was followed by increases in cyclin-dependent kinase inhibitors (CDKN) p27/CDKN1B and p21/CDKN1A that mediate cell cycle exit with differentiation. Effects were independent of the TP53, p16/CDKN2A and also the BRAF status of the melanoma cells. Resistance, when observed, was pharmacologic, characterized by diminished ability of decitabine to deplete DNMT1. Treatment of murine melanoma models in vivo with intermittent, low-dose decitabine, administered sub-cutaneously to limit high peak drug levels that cause cytotoxicity and increase exposure time for DNMT1 depletion, and with tetrahydrouridine to decrease decitabine metabolism and further increase exposure time, inhibited tumor growth and increased molecular and tumor stromal factors implicated in melanocyte differentiation. Modification of decitabine dose, schedule and formulation for differentiation rather than cytotoxic objectives inhibits the growth of melanoma cells in vitro and in vivo.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Azacitidina/análogos & derivados , DNA (Citosina-5-)-Metiltransferases/metabolismo , Epigênese Genética , Melanoma Experimental/tratamento farmacológico , Animais , Apoptose , Azacitidina/administração & dosagem , Azacitidina/farmacologia , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteínas Inibidoras de Quinase Dependente de Ciclina/biossíntese , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/análise , Metilação de DNA , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas B-raf/biossíntese , Fatores de Transcrição SOX9/biossíntese , Análise de Sequência de DNA , Tetra-Hidrouridina/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
11.
Invest Ophthalmol Vis Sci ; 52(8): 5529-35, 2011 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-21519029

RESUMO

PURPOSE: In contrast to many malignancies showing evidence that interleukin-1 (IL-1) promotes progression through effects on tumor vascularity and myeloid suppressor cell populations, in uveal melanoma there is evidence that IL-1 can inhibit progression. METHODS: The effects of the IL-1 receptor antagonist IL-1ra against the aggressive/invasive MUM2B and the nonaggressive/noninvasive OCM1 uveal melanoma models were examined in vitro and in vivo in mouse xenografts. Vascularity and myeloid suppressor cell populations and their regulators were assessed. RESULTS: In vitro, IL-1, and IL-1ra did not affect the proliferation of the uveal melanoma cells or their production of IL-1, IL-6, transforming growth factor (TGF) ß, or VEGF. In vivo, IL-1ra treatment resulted in substantial growth inhibition of MUM2B tumors; less inhibition was observed against OCM1 tumors. Periodic acid-Schiff loops and CD11b⁺ macrophages within the tumor stroma decreased in vivo; CD31⁺ blood vessels were not altered. IL-1ra treatment in vivo did not affect tumor-derived IL-1, IL-6, TGF-ß, or VEGF. In contrast, host IL-1ß, IL-6, and tumor necrosis factor decreased. Host VEGF was not altered. Intratumoral IL-12(p40) and CXCL10, markers of host M1 polarization, increased, and intratumoral arginase and CD206, markers of myeloid-derived suppressor cells (MDSC) and M2 macrophage polarization, decreased. IL-1ra treatment in vivo also reduced splenic CD11b⁺Gr1⁺ MDSC. CONCLUSIONS: IL-1 may play a role in promoting uveal melanoma progression. Inhibiting IL-1 with IL-1ra inhibits tumor growth in vivo but not in vitro. Tumor stroma is modified, myeloid suppressor cells are reduced, and M1 macrophage polarization is increased in vivo.


Assuntos
Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Melanoma/patologia , Neoplasias Uveais/patologia , Animais , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Citocinas/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Técnicas Imunoenzimáticas , Injeções Intraperitoneais , Masculino , Melanoma/genética , Melanoma/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/metabolismo , Células Estromais/patologia , Células Tumorais Cultivadas , Neoplasias Uveais/genética , Neoplasias Uveais/metabolismo
12.
Vaccine ; 28(50): 7837-43, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-20937315

RESUMO

Recombinant adeno-associated virus (rAAV) is being tested as a vaccine vector, but the cellular immune responses elicited in animal tumor models have not been completely protective. The adjuvant effects of the TLR7 agonist, imiquimod, and the TLR9 agonist, ODN1826, were tested with rAAV expressing the melanoma antigen, Trp2. Mice immunized with rAAV-TRP2 and either TLR agonist alone generated T-helper-1 antitumor immune responses. Antitumor activity in all experiments was still incomplete. Furthermore, antitumor activity was not achieved when the combination of ODN1826 and imiquimod was used as adjuvant. In vitro, the combination increased IL-10 production by dendritic cells. In vivo, the combination reduced T-helper-1 response and dendritic cell activation and increased myeloid suppressor cells; regulatory T cells were not significantly modulated. Depletion of myeloid derived suppressor cells enhanced the antitumor activity of immunization with rAAV-TRP2 and the imiquimod-ODN1826 combination; depletion of regulatory T cells did not. TLR7 and TLR9 agonists can be used to enhance the immune response to rAAV immunogens, but antagonism can be observed when combined. Suppressor mechanisms, including those mediated by myeloid cells, may negatively regulate the antitumor immune response.


Assuntos
Adjuvantes Imunológicos/farmacologia , Vacinas Anticâncer/imunologia , Melanoma Experimental/prevenção & controle , Proteínas de Membrana/imunologia , Fragmentos de Peptídeos/imunologia , Receptor 7 Toll-Like/agonistas , Receptor Toll-Like 9/agonistas , Aminoquinolinas/farmacologia , Animais , Células Cultivadas , DNA/farmacologia , Células Dendríticas/imunologia , Dependovirus/imunologia , Feminino , Imiquimode , Interleucina-10/imunologia , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Oligodesoxirribonucleotídeos , Linfócitos T Reguladores/imunologia
13.
Anticancer Res ; 30(2): 345-54, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20332438

RESUMO

BACKGROUND: Interleukin-1 (IL-1) has been implicated in the vascular and immune suppressor cell responses that promote tumor progression. IL-1, however, has also been shown to inhibit tumor progression by promoting antitumor immune responses and by enhancing the activity of chemotherapy. MATERIALS AND METHODS: The effects of IL-1 receptor antagonist (IL-1Ra), alone and combined with temozolomide and docetaxel chemotherapy, were examined in vitro and in vivo against microscopic and macroscopic mouse B16 melanoma. RESULTS: IL-1Ra did not demonstrate antitumor activity in vitro, alone or combined with chemotherapy. When administered in vivo at the time when B16 cells were inoculated, IL-1Ra inhibited tumor growth. It did not affect growth when administered after tumors had established and were macroscopic. IL-1Ra increased M1 macrophage polarization and reduced myeloid-derived suppressor cells in mice with macroscopic tumors. Regulatory T-cells, tumor vascularity, and tumor interstitial fluid pressure were not significantly altered. Pre-treatment but not concurrent treatment with IL-1Ra enhanced the antitumor activity of chemotherapy in vivo against macroscopic tumors. Whereas chemotherapy reduced myeloid suppressor cells systemically, chemotherapy increased markers of myeloid suppressor cells intratumorally. These effects were attenuated by pre-treatment with IL-1Ra. CONCLUSION: The effects of IL-1 and chemotherapy in melanoma are complex. IL-1Ra modifies myeloid suppressor cell populations and may have a role with chemotherapy in the treatment of established melanoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antirreumáticos/uso terapêutico , Proteína Antagonista do Receptor de Interleucina 1/uso terapêutico , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Animais , Western Blotting , Proliferação de Células , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Docetaxel , Quimioterapia Combinada , Líquido Extracelular/efeitos dos fármacos , Feminino , Citometria de Fluxo , Técnicas Imunoenzimáticas , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/imunologia , Taxoides/administração & dosagem , Temozolomida , Células Tumorais Cultivadas
14.
Melanoma Res ; 18(6): 420-30, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18971787

RESUMO

Uveal melanoma is refractory to chemotherapy. The receptor tyrosine kinase inhibitor, imatinib mesylate, has demonstrated antiproliferative effects against uveal melanoma cells in vitro. The effects of imatinib mesylate, alone and combined with the alklyating agent, temozolomide, were examined in vivo as well as in vitro. Proliferation and angiogenic factor production of human uveal melanoma cell lines in response to imatinib mesylate and temozolomide were examined in vitro. Tumor growth, angiogenic factor production, tumor interstitial fluid pressure, and stroma constituents in response to imatinib mesylate and temozolomide were examined in vivo in mice bearing human uveal melanoma xenografts. Imatinib mesylate in vitro antagonized the antiproliferative effects of temozolomide and increased the production of angiogenic factors. In contrast, pretreatment with imatinib mesylate in vivo could improve the antitumor activity of temozolomide. Imatinib mesylate in vivo decreased the production of angiogenic factors in the tumor stroma and tumor interstitial fluid pressure. These effects were transient. Increases in angiogenic factors, interstitial fluid pressure, and tumor infiltrating macrophages were observed with continued imatinib mesylate treatment in vivo. The antitumor effects of imatinib mesylate can vary in vivo when compared with in vitro. Imatinib mesylate can both positively and negatively modify host-tumor interactions in uveal melanoma.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Melanoma/metabolismo , Piperazinas/farmacologia , Pirimidinas/farmacologia , Neoplasias Uveais/metabolismo , Animais , Antineoplásicos Alquilantes/farmacologia , Benzamidas , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Humanos , Mesilato de Imatinib , Camundongos , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Clin Cancer Res ; 12(9): 2862-8, 2006 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-16675582

RESUMO

PURPOSE: Bisphosphonates (such as risedronate and zoledronate) are widely used inhibitors of bone resorption. Despite their in vitro antiproliferative effects in various cancer cells, bisphosphonates have not exhibited significant antitumor efficacy in animal models of visceral cancer, which may be due to their poor bioavailability. The diagnostic use of radioactive bisphosphonates has revealed the accumulation of bisphosphonates in mesothelioma, which prompted us to test the antitumor efficacy of bisphosphonates in this disease. EXPERIMENTAL DESIGN AND RESULTS: Treatment with either risedronate or zoledronate (2 x 10(-4) to 2 x 10(-6) mol/L) inhibited the growth of AB12 and AC29 mouse mesothelioma cells and induced the accumulation of unprenylated Rap1A in these cells. Both these in vitro effects were reversed by geranygeraniol, an end product of the mevalonate pathway that these bisphosphonates inhibit. Both bisphosphonates also induced the phosphorylation of the p38 mitogen-activated protein kinase in AB12 and AC29 cells. The inhibition of p38 augmented bisphosphonate-induced growth inhibition in these cells. Bisphosphonate-induced p38 phosphorylation was not reversible by geranylgeraniol. Risedronate (15 mg/kg) and zoledronate (0.5 mg/kg) inhibited the growth of s.c. tumors and increased the median survival of mice with i.p. mesothelioma tumors in vivo. DISCUSSION: In conclusion, risedronate and zoledronate inhibit the mevalonate pathway and induce p38 activation in mesothelioma cells in vitro. The effects on the mevalonate pathway dominate because the net result is growth inhibition. Both bisphosphonates also inhibit mesothelioma tumor growth in vivo and prolong the survival of mesothelioma-bearing mice. These results support further study of bisphosphonates in the management of mesothelioma.


Assuntos
Difosfonatos/uso terapêutico , Mesotelioma/patologia , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Difosfonatos/farmacologia , Modelos Animais de Doenças , Ácido Etidrônico/análogos & derivados , Ácido Etidrônico/uso terapêutico , Imidazóis/uso terapêutico , Mesotelioma/tratamento farmacológico , Camundongos , Fosforilação , Ácido Risedrônico , Ácido Zoledrônico
16.
Cancer Biother Radiopharm ; 21(6): 553-60, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17257070

RESUMO

PURPOSE: Antibodies targeting GD3 gangliosides highly expressed on melanomas mediate immune effector functions in vitro and inhibit animal model melanoma tumor growth in vivo. Because GD3 is expressed also on a subpopulation of human lymphocytes, we characterized the in vitro immune effects of murine R24 and a chimeric anti-GD3 antibody (KW-2871). DESIGN: Anti-GD3 complement-mediated (CMC) and antibody-dependent cellular cytotoxicity (ADCC) were tested against cell line Mel-624. Antibody-mediated lymphocyte expression of interleukin (IL)-2, IL-4, IL-10, and interferon-gamma (IFN-gamma) was quantified. The effect of antibody and antibody-treated lymphocyte supernates on effector cell ADCC and Fc receptor expression were evaluated. RESULTS: R24 and KW-2871 antibodies mediated CMC and ADCC to the Mel-624 cell line. R24 induced potent lymphocyte proliferation and enhanced lymphocyte RNA expression of IL-4 (2-4 logs), IL-10, and IFN-gamma (> 10-fold). KW-2871 induced no lymphocyte proliferation and had minimal effects on lymphokine expression (< 5-fold). Preincubation of effector cells with either antibody inhibited ADCC and reduced monocyte expression of FcgammaRI and II. Supernates of effector cells preincubated with either antibody were able to inhibit ADCC. CONCLUSIONS: R24 and KW-2871 antibody differ in their lymphocyte proliferation and lymphokine release activity but have similar inhibition of lymphocyte ADCC and FcgammaR expression in vitro.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/toxicidade , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Gangliosídeos/imunologia , Linfócitos T/imunologia , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Citocinas/metabolismo , Humanos , RNA Mensageiro/genética , Receptores Fc/imunologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
17.
Breast Cancer Res Treat ; 93(2): 169-75, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16187237

RESUMO

We previously identified a CD2-initiated signaling pathway which inhibits activation-induced cell death in mitogen-stimulated human gammadelta-T cells permitting the large-scale expansion of these cells. Here we report the innate anti-tumor activity of expanded human gammadelta-T cells against human breast cancer cells. Apoptosis-resistant human gammadelta-T cells which were expanded in vitro from cultured human peripheral blood mononuclear cells displayed lytic activity against breast cancer cell lines MDA-MB-231, MCF-7 and T-47D, but failed to kill normal human skin fibroblasts and normal human liver cells. Monoclonal antibodies (mAb) directed against the gammadelta-T cell receptor (TCR) or mAb directed against either the Vgamma9 or the Vdelta2 TCR chains were able to block gammadelta-T cell-mediated lysis of MDA-MB-231 cells. In addition, mAb against intercellular adhesion molecules-1 (ICAM-1/CD54) or CD18 (beta subunit of ICAM-1 counter-receptor) also blocked gammadelta-T cell-mediated killing of MDA-MB-231 cells. Ex vivo expanded human gammadelta-T cells are thus able to innately recognize and kill human breast cancer cells in a gammadelta-TCR-dependent manner; ICAM-1 and CD18 also appear to be involved in the interactions between sensitive breast cancer cells and cytolytic gammadelta-T cells. As apoptosis-resistant human gammadelta-T cells can now readily be expanded to large numbers (clinical scale), these findings must be considered in the context of developing adoptive immunotherapy strategies to exploit gammadelta-T cell innate immune responses for the primary or adjuvant treatment of breast cancer.


Assuntos
Apoptose/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Linfócitos T/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Testes Imunológicos de Citotoxicidade , Citotoxicidade Imunológica/imunologia , Feminino , Humanos
18.
J Immunother ; 28(4): 382-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16000957

RESUMO

Immunization with plasmid DNA represents an attractive method for increasing cellular immune responses against cancer antigens. The safety and immunologic response of a plasmid encoding the MART-1 melanocyte differentiation antigen was evaluated in 12 patients with resected melanoma at risk for relapse. As a control, patients were also administered a plasmid encoding hepatitis B surface antigen (HBsAg). After establishing immunologic activity of the vaccines in mice, groups of three to six HLA-A2-positive patients were enrolled into one of three cohorts in which they received intramuscular injections of the MART-1 plasmid into the right deltoid and the HBsAg plasmid into the left deltoid at doses of 0.1, 0.3, or 1.0 mg on days 1, 43, 85, and 127. Injections were well tolerated. Toxicity was limited to grade 1 pain and injection site tenderness. Systemic toxicity was not observed. Although baseline MART-1-specific lymphoproliferative and ELISPOT responses were evident, no patient manifested increases after injection of the MART-1 plasmid. Furthermore, changes in MART-1-specific precursors were not evident after immunization as assessed by an in vitro stimulation assay. No patients manifested a lymphoproliferative response to HBsAg antigen, and significant antibody responses to HBsAg were also not observed. Although injections were safe, the authors could not show significant immunologic responses to plasmid encoding MART-1 or HBsAg using the dose, schedule, and route of administration applied. This study underscores species differences in the ability to respond to plasmid immunogens.


Assuntos
Melanoma/prevenção & controle , Proteínas de Neoplasias/imunologia , Vacinas de DNA/imunologia , Adulto , Idoso , Animais , Formação de Anticorpos/imunologia , Antígenos de Neoplasias , Proliferação de Células , Feminino , Antígenos de Superfície da Hepatite B/genética , Antígenos de Superfície da Hepatite B/imunologia , Humanos , Imunidade Celular/imunologia , Interferon gama/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Ativação Linfocitária/imunologia , Antígeno MART-1 , Masculino , Melanoma/imunologia , Melanoma/cirurgia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Recidiva Local de Neoplasia/prevenção & controle , Plasmídeos/administração & dosagem , Plasmídeos/imunologia , Especificidade da Espécie , Vacinação , Vacinas de DNA/administração & dosagem
19.
Cancer Biol Ther ; 4(3): 289-94, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15753654

RESUMO

Dendritic cells (DCs) are a central element in the development of antigen-specific immune responses. The lack of a specific and efficient technique for the in vivo delivery of antigens to DCs remains a major obstacle limiting a vaccine's ability to induce an effective immune response. The efficacy of adenoviral (Ad) vectors in this regard can be enhanced through alterations in vector tropism such that DC-targeted transduction is achieved. Here, the efficiency of DC transduction by Ad vectors retargeted to DC-specific ICAM-3 grabbing nonintegrin (DC-SIGN) was studied and compared to that of Ad vectors retargeted through CD40. A comparable and significant enhancement of gene transfer to monocyte derived DCs (MDDCs) was accomplished by means of an Ad vector harboring the Fc-binding domain of Staphylococcus aureus protein A in combination with antibodies to DC-SIGN or to CD40 or with fused complexes of human Ig-Fc with their natural ligands, i.e., ICAM-3 or CD40L, respectively. Whereas CD40-targeted Ad transduction resulted in a more profound phenotypic DC maturation, DC-SIGN- and CD40-targeted Ad both induced similar levels of IL-12 secretion. These data demonstrate the usefulness of DC-SIGN as a DC-restricted targeting motif for Ad-mediated vaccination strategies.


Assuntos
Adenoviridae/genética , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/imunologia , Vetores Genéticos/genética , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução Genética/métodos , Antígenos CD40/genética , Vacinas Anticâncer/genética , Linhagem Celular , Humanos , Imunoterapia Adotiva , Monócitos/imunologia
20.
Int J Cancer ; 113(3): 406-14, 2005 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-15455351

RESUMO

Deficiency in costimulatory molecule expression has been implicated in the ability of tumors to escape immune effectors. The activity of the intratumoral administration of recombinant fowlpox vectors expressing a triad of costimulatory molecules (rF-TRICOM) was evaluated in the asbestos-induced AB12 and AC29 mouse models of mesothelioma. Mesothelioma cell infected with rF-TRICOM expressed high levels of the costimulatory molecules. Prolongation of survival was observed in mice receiving rF-TRICOM in AB12 and AC29 intraperitoneal models. Complete tumor regressions were observed in mice receiving intratumoral rF-TRICOM in the AB12 subcutaneous tumor model. Tumor regressions were associated with the development of serum IgG reactivities to mesothelioma-associated determinants and specific systemic cytolytic activity, and responding mice were capable of rejecting tumors upon re-challenge. Antitumor activity was also observed in mice with established AB12 tumor vaccinated with irradiated rF-TRICOM-infected AB12 cells. The antitumor activity of intratumoral rF-TRICOM was superior to that of the intratumoral injection of a fowlpox vector expressing granulocyte-macrophage colony stimulating factor (rF-GM-CSF). AB12 and AC29 tumors were found to produce GM-CSF and to have substantial macrophage infiltration. Production of GM-CSF decreased in vivo in tumors injected with rF-TRICOM. rF-TRICOM and wild-type fowlpox inhibited the growth of AB12 and AC29 cells in vitro; less inhibition was observed with rF-GM-CSF. These results indicate that the intratumoral injection of rF-TRICOM has significant activity in mouse models of mesothelioma and can elicit a systemic antitumor immune response. The results also suggest potential limitations to the intratumoral administration of cytokines, such as GM-CSF, in mesothelioma.


Assuntos
Varíola Aviária/genética , Vetores Genéticos/uso terapêutico , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Mesotelioma/terapia , Vacinas Sintéticas/uso terapêutico , Animais , Amianto/toxicidade , Aves , Vacinas Anticâncer , Carcinógenos/toxicidade , Proliferação de Células/efeitos dos fármacos , DNA Recombinante , Feminino , Varíola Aviária/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Imunoglobulina G/sangue , Mesotelioma/induzido quimicamente , Mesotelioma/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos CBA , Taxa de Sobrevida , Vacinação , Vacinas Sintéticas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA