Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncoimmunology ; 5(2): e1075693, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27057448

RESUMO

Glioblastoma multiforme (GBM) is a highly malignant tumor with a poor outcome that is often positive for human cytomegalovirus (HCMV). GBM patients often have excessive numbers of neutrophils and macrophages near and within the tumor. Here, we characterized the cytokine patterns in the blood of GBM patients with and without Valganciclovir treatment. Furthermore, we determined whether neutrophil activation is related to HCMV status and patient outcome. Blood samples for analyses of cytokines and growth factors were collected from 42 GBM patients at the time of diagnosis (n = 42) and at weeks 12 and 24 after surgery. Blood neutrophils of 28 GBM patients were examined for CD11b expression. The levels of pro- and anti-inflammatory cytokines and chemokines-including interleukin (IL)-1ß, IL-2, IL-6, IL-8, IL-10, IL-12p70, IL-17A, transforming growth factor (TGF)-ß1, interferon-γ, interferon-α, tumor necrosis factor α, and monocyte chemoattractant protein (MCP)-1were analyzed with a bead-based flow cytometry assay. During the first six months after surgery, neutrophil activity was increased in 12 patients and was unchanged or decreased in 16. Patients with increased neutrophil activity had enhanced IL-12p70, high grade HCMV and a shorter time to tumor progression (TTP) than patients without or decreased neutrophil activity (median TTP; 5.4 vs. 12 months, 95% confidence interval; 1.6-10 vs. 0.1-0.6, hazard ratio = 3 vs. 0.4, p = 0.004). The levels of IL-12p70 were significantly decreased in Valganciclovir treated patients (n = 22, T 12W vs. T 24W, p = 0.03). In conclusion, our findings suggest that neutrophil activation is an early sign of tumor progression in GBM patients.

2.
BMJ Case Rep ; 20152015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26670887

RESUMO

Nearly all glioblastomas (GBMs), brain tumours with very poor prognosis, are infected with human cytomegalovirus (CMV). The anti-CMV drug valganciclovir (VGCV) has shown promise as a treatment option for patients with GBM, but its penetration into the central nervous system (CNS) is unknown. Here we describe a patient with GMB receiving VGCV in whom an intracerebral microdialysis catheter was implanted and ganciclovir (GCV) concentrations in brain extracellular fluid (BECF) and serum were monitored. GCV was rapidly absorbed. Cmax values (at 3 h) in serum and BECF were 19.6 and 10.2 µmol/L, T½ values were 3.2 and 4.5 h, and plasma and BECF AUC0-∞ values were 90.7 and 75.9 µmol h/L, respectively. Thus, VGCV treatment results in significant intracerebral levels of GCV that may be sufficient for therapeutic effects. Further studies of this drug in patients with GBM are warranted.


Assuntos
Antivirais/farmacocinética , Neoplasias Encefálicas/metabolismo , Ganciclovir/análogos & derivados , Ganciclovir/farmacocinética , Glioblastoma/metabolismo , Antivirais/administração & dosagem , Encéfalo/metabolismo , Neoplasias Encefálicas/virologia , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/metabolismo , Monitoramento de Medicamentos , Espaço Extracelular/metabolismo , Seguimentos , Ganciclovir/administração & dosagem , Glioblastoma/virologia , Humanos , Masculino , Microdiálise , Pessoa de Meia-Idade , Resultado do Tratamento , Valganciclovir
3.
Oncoimmunology ; 4(9): e1036211, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26405601

RESUMO

Patients with glioblastoma multiforme (GBM) are immunosuppressed and have a broad range of immunological defects in both innate and adaptive immune responses. GBMs are frequently infected with human cytomegalovirus (HCMV), a virus capable of causing immunosuppression. In 42 HCMV-positive GBM patients in a clinical trial (VIGAS), we investigated T-cell phenotypes in the blood and assessed their relation to survival. Blood was collected before and 3, 12, and 24 weeks after surgery, and the frequency of T-cell subsets was compared with that in 26 age-matched healthy controls. GBM patients had lower levels of CD3 cells than the controls, but had significantly higher levels of CD4+CD28- T cells before and 3 and 12 weeks after surgery and increased levels of CD4+CD57+ and CD4+CD57+CD28+ T cells at all-time points. These T-cell subsets were associated with both immunosenescence and HCMV infection. GBM patients also had higher levels of γδ T cells at all-times after surgery and lower levels of CD4+CD25+ cells before and 3 weeks after surgery than healthy controls. Overall survival was significantly shorter in patients with higher levels of CD4+CD28- T cells (p = 0.025), CD4+CD57+ T (p = 0.025) cells, and CD4+CD28-CD57+CD28- T cells (p < 0.0004) at 3 weeks after surgery. Our findings indicate that signs of immunosenescence in the CD4+ compartment are associated with poor prognosis in patients with HCMV-positive GBMs and may reflect the HCMV activity in their tumors.

4.
Int J Cancer ; 133(10): 2351-61, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23661597

RESUMO

Neuroblastoma is the most common and deadly tumor of childhood, where new therapy options for patients with high-risk disease are highly warranted. Human cytomegalovirus (HCMV) is prevalent in the human population and has recently been implicated in different cancer forms where it may provide mechanisms for oncogenic transformation, oncomodulation and tumor cell immune evasion. Here we show that the majority of primary neuroblastomas and neuroblastoma cell lines are infected with HCMV. Our analysis show that HCMV immediate-early protein was expressed in 100% of 36 primary neuroblastoma samples, and HCMV late protein was expressed in 92%. However, no infectious virus was detected in primary neuroblastoma tissue extracts. Remarkably, all six human neuroblastoma cell lines investigated contained CMV DNA and expressed HCMV proteins. HCMV proteins were expressed in neuroblastoma cells expressing the proposed stem cell markers CD133 and CD44. When engrafted into NMRI nu/nu mice, human neuroblastoma cells expressed HCMV DNA, RNA and proteins but did not produce infectious virus. The HCMV-specific antiviral drug valganciclovir significantly reduced viral protein expression and cell growth both in vitro and in vivo. These findings indicate that HCMV is important for the pathogenesis of neuroblastoma and that anti-viral therapy may be a novel adjuvant treatment option for children with neuroblastoma.


Assuntos
Infecções por Citomegalovirus/complicações , Citomegalovirus/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neuroblastoma/virologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Criança , Pré-Escolar , Infecções por Citomegalovirus/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Feminino , Ganciclovir/farmacologia , Ganciclovir/uso terapêutico , Glicoproteínas/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Nus , Neuroblastoma/metabolismo , Peptídeos/metabolismo , Prevalência , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Clin Virol ; 57(1): 36-42, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23391370

RESUMO

BACKGROUND: Patients with glioblastoma multiforme (GBM) generally live 12-15 months after diagnosis, despite maximal surgical resection, adjuvant radiotherapy, and chemotherapy. HCMV has been detected in 90-100% of GBMs. We recently found that low grade HCMV infection in GBM tumours was highly associated with survival over 18 months (case-control study). Here, we sought to determine whether low-grade HCMV infection in GBMs is associated with prolonged survival in a consecutive patient cohort, analysed retrospectively. STUDY DESIGN: Tumour samples from 75 consecutive GBM patients treated surgically at Karolinska University Hospital in 2004-2005 were examined by immunohistochemistry (IHC) and in situ hybridization for HCMV proteins and DNA, respectively. Tumours were graded 1-4, depending on the percentage of positive cells by IHC. Low-grade HCMV was defined as grade 1 (< 25% of HCMV infected tumour cells). Time to tumour progression (TTP) and survival data were analysed with Cox regression and Kaplan-Meier models. RESULTS: HCMV infection was detected in 74 of 75 tumours (99%). In patients with low-grade HCMV infection, median survival was 20 months longer than in patients with high-grade infections (P = 0.036, HR: 2.2), and TTP was 8 months longer (P = 0.1, HR: 1.8). Two-year survival was much higher in patients with low-grade HCMV infection (63.6% vs. 17.2%, P = 0.003). CONCLUSION: The longer survival in patients whose tumours had low-grade HCMV infection suggests that the level of HCMV infection in GBMs has a prognostic value and that HCMV may contribute to the pathogenesis of GBM.


Assuntos
Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/complicações , Citomegalovirus/isolamento & purificação , Glioblastoma/virologia , Neoplasias Encefálicas/diagnóstico , Progressão da Doença , Feminino , Glioblastoma/diagnóstico , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico
6.
J Clin Invest ; 121(10): 4043-55, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21946257

RESUMO

Medulloblastomas are the most common malignant brain tumors in children. They express high levels of COX-2 and produce PGE2, which stimulates tumor cell proliferation. Human cytomegalovirus (HCMV) is prevalent in the human population and encodes proteins that provide immune evasion strategies and promote oncogenic transformation and oncomodulation. In particular, HCMV induces COX-2 expression; STAT3 phosphorylation; production of PGE2, vascular endothelial growth factor, and IL-6; and tumor formation in vivo. Here, we show that a large proportion of primary medulloblastomas and medulloblastoma cell lines are infected with HCMV and that COX-2 expression, along with PGE2 levels, in tumors is directly modulated by the virus. Our analysis indicated that both HCMV immediate-early proteins and late proteins are expressed in the majority of primary medulloblastomas. Remarkably, all of the human medulloblastoma cell lines that we analyzed contained HCMV DNA and RNA and expressed HCMV proteins at various levels in vitro. When engrafted into immunocompromised mice, human medulloblastoma cells induced expression of HCMV proteins. HCMV and COX-2 expression correlated in primary tumors, cell lines, and medulloblastoma xenografts. The antiviral drug valganciclovir and the specific COX-2 inhibitor celecoxib prevented HCMV replication in vitro and inhibited PGE2 production and reduced medulloblastoma tumor cell growth both in vitro and in vivo. Ganciclovir did not affect the growth of HCMV-negative tumor cell lines. These findings imply an important role for HCMV in medulloblastoma and suggest HCMV as a novel therapeutic target for this tumor.


Assuntos
Neoplasias Encefálicas/virologia , Citomegalovirus/isolamento & purificação , Meduloblastoma/virologia , Adulto , Animais , Antivirais/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Celecoxib , Linhagem Celular Tumoral , Criança , Pré-Escolar , Inibidores de Ciclo-Oxigenase 2/farmacologia , Citomegalovirus/efeitos dos fármacos , Citomegalovirus/patogenicidade , Citomegalovirus/fisiologia , Dinoprostona/biossíntese , Feminino , Ganciclovir/análogos & derivados , Ganciclovir/farmacologia , Humanos , Lactente , Masculino , Meduloblastoma/tratamento farmacológico , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Valganciclovir , Replicação Viral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
7.
Stem Cell Res ; 2(1): 56-67, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19383409

RESUMO

Transplantation of human neural stem cells (NSCs) and their derivatives is a promising future treatment for neurodegenerative disease and traumatic nervous system lesions. An important issue is what kind of immunological reaction the cellular transplant and host interaction will result in. Previously, we reported that human NSCs, despite expressing MHC class I and class II molecules, do not trigger an allogeneic T cell response. Here, the immunocompetence of human NSCs, as well as differentiated neural cells, was further studied. Astrocytes expressed both MHC class I and class II molecules to a degree equivalent to that of the NSCs, whereas neurons expressed only MHC class I molecules. Neither the NSCs nor the differentiated cells triggered an allogeneic lymphocyte response. Instead, these potential donor NSCs and astrocytes, but not the neurons, exhibited a suppressive effect on an allogeneic immune response. The suppressive effect mediated by NSCs most likely involves cell-cell interaction. When the immunogenicity of human NSCs was tested in an acute spinal cord injury model in rodent, a xenogeneic rejection response was triggered. Thus, human NSCs and their derived astrocytes do not initiate, but instead suppress, an allogeneic response, while they cannot block a graft rejection in a xenogeneic setting.


Assuntos
Astrócitos/transplante , Neurônios/transplante , Transplante de Células-Tronco , Linfócitos T/imunologia , Animais , Astrócitos/imunologia , Modelos Animais de Doenças , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II , Humanos , Neurônios/imunologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal , Células-Tronco/imunologia , Transplante Heterólogo/imunologia , Transplante Homólogo/imunologia
8.
J Natl Cancer Inst ; 101(7): 488-97, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19318640

RESUMO

BACKGROUND: The mechanism by which human cytomegalovirus (HCMV) stimulates oncogenesis is unclear. Because cellular immortalization and transformation require telomerase activation by expression of the telomerase reverse transcriptase (hTERT) gene, we examined the role of HCMV in telomerase activation. METHODS: Normal human diploid fibroblasts (HDFs) and human malignant glioma (MG) cell lines were infected with HCMV or transfected with expression vectors encoding HCMV immediate early (IE) antigen 72 or 86. hTERT expression and promoter activity and telomerase activity were evaluated using reverse transcription-polymerase chain reaction, a luciferase reporter assay, and a telomeric repeat amplification protocol, respectively. hTERT promoter occupancy by the transcription factor Sp1, IE antigens, and histone deacetylases (HDACs) was assessed by chromatin immunoprecipitation. hTERT and IE protein expression in human primary glioblastoma multiforme (GBM) was determined immunohistochemically. All statistical tests were two-sided. RESULTS: In telomerase and hTERT-negative HDFs, HCMV infection induced constitutive hTERT expression and telomerase activation. The hTERT promoter activity in HDFs and MG cell lines was statistically significantly enhanced by HCMV in a dose-dependent manner (mean luciferase activity [arbitrary units] in control HDFs and in HDFs infected with HCMV at multiplicities of infection [MOIs] of 0.1 = 6 and 521, respectively, difference = 515, 95% CI = 178 to 850; mean activity at MOI of 1 and 10 = 8828 and 59,923, respectively; P < .001 comparing control with HCMV-infected cells at all MOIs). Ectopic expression of HCMV IE-72 protein also stimulated hTERT promoter activity in HDFs. HCMV-mediated transactivation of the hTERT gene was dependent on the presence of Sp1-binding sites in the hTERT promoter and was accompanied by increases in Sp1 binding, acetylation of histone H3, and a reduction in HDAC binding at the core promoter. In specimens of GBM, HCMV IE and hTERT proteins were colocalized in malignant cells and their levels paralleled each other. CONCLUSIONS: HCMV activates telomerase in both HDFs and malignant cells. These findings begin to reveal a novel mechanism by which HCMV infection may be linked to or modulate oncogenesis through telomerase activation.


Assuntos
Infecções por Citomegalovirus/metabolismo , Citomegalovirus/metabolismo , Fibroblastos/metabolismo , Glioma/enzimologia , Telomerase/metabolismo , Telômero/metabolismo , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Infecções por Citomegalovirus/enzimologia , Ativação Enzimática , Fibroblastos/enzimologia , Imunofluorescência , Glioblastoma/metabolismo , Glioma/metabolismo , Glioma/virologia , Humanos , Immunoblotting , Imuno-Histoquímica , Luciferases/metabolismo , Vírus Oncogênicos/metabolismo , RNA Mensageiro/metabolismo , Projetos de Pesquisa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Telomerase/genética , Telômero/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA