Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
JAMA Oncol ; 9(1): 112-121, 2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36394838

RESUMO

Importance: Glioblastoma is the most lethal primary brain cancer. Clinical outcomes for glioblastoma remain poor, and new treatments are needed. Objective: To investigate whether adding autologous tumor lysate-loaded dendritic cell vaccine (DCVax-L) to standard of care (SOC) extends survival among patients with glioblastoma. Design, Setting, and Participants: This phase 3, prospective, externally controlled nonrandomized trial compared overall survival (OS) in patients with newly diagnosed glioblastoma (nGBM) and recurrent glioblastoma (rGBM) treated with DCVax-L plus SOC vs contemporaneous matched external control patients treated with SOC. This international, multicenter trial was conducted at 94 sites in 4 countries from August 2007 to November 2015. Data analysis was conducted from October 2020 to September 2021. Interventions: The active treatment was DCVax-L plus SOC temozolomide. The nGBM external control patients received SOC temozolomide and placebo; the rGBM external controls received approved rGBM therapies. Main Outcomes and Measures: The primary and secondary end points compared overall survival (OS) in nGBM and rGBM, respectively, with contemporaneous matched external control populations from the control groups of other formal randomized clinical trials. Results: A total of 331 patients were enrolled in the trial, with 232 randomized to the DCVax-L group and 99 to the placebo group. Median OS (mOS) for the 232 patients with nGBM receiving DCVax-L was 19.3 (95% CI, 17.5-21.3) months from randomization (22.4 months from surgery) vs 16.5 (95% CI, 16.0-17.5) months from randomization in control patients (HR = 0.80; 98% CI, 0.00-0.94; P = .002). Survival at 48 months from randomization was 15.7% vs 9.9%, and at 60 months, it was 13.0% vs 5.7%. For 64 patients with rGBM receiving DCVax-L, mOS was 13.2 (95% CI, 9.7-16.8) months from relapse vs 7.8 (95% CI, 7.2-8.2) months among control patients (HR, 0.58; 98% CI, 0.00-0.76; P < .001). Survival at 24 and 30 months after recurrence was 20.7% vs 9.6% and 11.1% vs 5.1%, respectively. Survival was improved in patients with nGBM with methylated MGMT receiving DCVax-L compared with external control patients (HR, 0.74; 98% CI, 0.55-1.00; P = .03). Conclusions and Relevance: In this study, adding DCVax-L to SOC resulted in clinically meaningful and statistically significant extension of survival for patients with both nGBM and rGBM compared with contemporaneous, matched external controls who received SOC alone. Trial Registration: ClinicalTrials.gov Identifier: NCT00045968.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Temozolomida/uso terapêutico , Estudos Prospectivos , Neoplasias Encefálicas/patologia , Recidiva , Células Dendríticas/patologia , Vacinação
2.
iScience ; 25(6): 104395, 2022 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-35637733

RESUMO

Oncolytic viruses (OVs) are emerging cancer immunotherapy. Despite notable successes in the treatment of some tumors, OV therapy for central nervous system cancers has failed to show efficacy. We used an ex vivo tumor model developed from human glioblastoma tissue to evaluate the infiltration of herpes simplex OV rQNestin (oHSV-1) into glioblastoma tumors. We next leveraged our data to develop a computational, model of glioblastoma dynamics that accounts for cellular interactions within the tumor. Using our computational model, we found that low stromal density was highly predictive of oHSV-1 therapeutic success, suggesting that the efficacy of oHSV-1 in glioblastoma may be determined by stromal-to-tumor cell regional density. We validated these findings in heterogenous patient samples from brain metastatic adenocarcinoma. Our integrated modeling strategy can be applied to suggest mechanisms of therapeutic responses for central nervous system cancers and to facilitate the successful translation of OVs into the clinic.

3.
J Clin Invest ; 129(4): 1671-1683, 2019 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-30855281

RESUMO

Cytomegalovirus (CMV) has been implicated in glioblastoma (GBM); however, a mechanistic connection in vivo has not been established. The purpose of this study is to characterize the effects of murine CMV (MCMV) on GBM growth in murine models. Syngeneic GBM models were established in mice perinatally infected with MCMV. We found that tumor growth was markedly enhanced in MCMV+ mice, with a significant reduction in overall survival compared with that of controls (P < 0.001). We observed increased angiogenesis and tumor blood flow in MCMV+ mice. MCMV reactivation was observed in intratumoral perivascular pericytes and tumor cells in mouse and human GBM specimens, and pericyte coverage of tumor vasculature was strikingly augmented in MCMV+ mice. We identified PDGF-D as a CMV-induced factor essential for pericyte recruitment, angiogenesis, and tumor growth. The antiviral drug cidofovir improved survival in MCMV+ mice, inhibiting MCMV reactivation, PDGF-D expression, pericyte recruitment, and tumor angiogenesis. These data show that MCMV potentiates GBM growth in vivo by increased pericyte recruitment and angiogenesis due to alterations in the secretome of CMV-infected cells. Our model provides evidence for a role of CMV in GBM growth and supports the application of antiviral approaches for GBM therapy.


Assuntos
Infecções por Citomegalovirus , Citomegalovirus/metabolismo , Glioblastoma , Neoplasias Experimentais , Neovascularização Patológica , Pericitos , Animais , Linhagem Celular Tumoral , Infecções por Citomegalovirus/metabolismo , Infecções por Citomegalovirus/patologia , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Linfocinas/metabolismo , Camundongos , Células NIH 3T3 , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/irrigação sanguínea , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Experimentais/virologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/virologia , Pericitos/metabolismo , Pericitos/patologia , Fator de Crescimento Derivado de Plaquetas/metabolismo
4.
Neurosurgery ; 84(3): E168-E170, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30629225

RESUMO

TARGET POPULATION: These recommendations apply to adult patients with new or recurrent solitary or multiple brain metastases from solid tumors as detailed in each section. QUESTION 1: Should patients with newly diagnosed metastatic brain tumors undergo stereotactic radiosurgery (SRS) compared with other treatment modalities? RECOMMENDATIONS: Level 3: SRS is recommended as an alternative to surgical resection in solitary metastases when surgical resection is likely to induce new neurological deficits, and tumor volume and location are not likely to be associated with radiation-induced injury to surrounding structures. Level 3: SRS should be considered as a valid adjunctive therapy to supportive palliative care for some patients with brain metastases when it might be reasonably expected to relieve focal symptoms and improve functional quality of life in the short term if this is consistent with the overall goals of the patient. QUESTION 2: What is the role of SRS after open surgical resection of brain metastasis? RECOMMENDATION: Level 3: After open surgical resection of a solitary brain metastasis, SRS should be used to decrease local recurrence rates. QUESTION 3: What is the role of SRS alone in the management of patients with 1 to 4 brain metastases? RECOMMENDATIONS: Level 3: For patients with solitary brain metastasis, SRS should be given to decrease the risk of local progression. Level 3: For patients with 2 to 4 brain metastases, SRS is recommended for local tumor control, instead of whole brain radiotherapy, when their cumulative volume is < 7 mL. QUESTION 4: What is the role of SRS alone in the management of patients with more than 4 brain metastases? RECOMMENDATION: Level 3: The use of stereotactic radiosurgery alone is recommended to improve median overall survival for patients with more than 4 metastases having a cumulative volume < 7 mL. The full guideline can be found at: https://www.cns.org/guidelines/guidelines-treatment-adults-metastatic-brain-tumors/chapter_4.


Assuntos
Neoplasias Encefálicas/radioterapia , Neurocirurgiões/normas , Guias de Prática Clínica como Assunto/normas , Radiocirurgia/normas , Adulto , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/secundário , Congressos como Assunto/normas , Progressão da Doença , Feminino , Humanos , Masculino , Qualidade de Vida , Radiocirurgia/mortalidade , Carga Tumoral
5.
Nat Commun ; 9(1): 4627, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30401823

RESUMO

Optimal treatment of brain metastases is often hindered by limitations in diagnostic capabilities. To meet this challenge, here we profile DNA methylomes of the three most frequent types of brain metastases: melanoma, breast, and lung cancers (n = 96). Using supervised machine learning and integration of DNA methylomes from normal, primary, and metastatic tumor specimens (n = 1860), we unravel epigenetic signatures specific to each type of metastatic brain tumor and constructed a three-step DNA methylation-based classifier (BrainMETH) that categorizes brain metastases according to the tissue of origin and therapeutically relevant subtypes. BrainMETH predictions are supported by routine histopathologic evaluation. We further characterize and validate the most predictive genomic regions in a large cohort of brain tumors (n = 165) using quantitative-methylation-specific PCR. Our study highlights the importance of brain tumor-defining epigenetic alterations, which can be utilized to further develop DNA methylation profiling as a critical tool in the histomolecular stratification of patients with brain metastases.


Assuntos
Neoplasias Encefálicas/genética , Metilação de DNA , Epigênese Genética , Epigenômica/métodos , Metástase Neoplásica/genética , Algoritmos , Neoplasias Encefálicas/patologia , DNA de Neoplasias , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/genética , Melanoma , Neoplasias Cutâneas , Aprendizado de Máquina Supervisionado , Melanoma Maligno Cutâneo
6.
Sci Data ; 5: 180245, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30398472

RESUMO

Brain metastases (BM) are one the most lethal and poorly managed clinical complications in cancer patients. These secondary tumors represent the most common intracranial neoplasm in adults, most frequently originating from lung cancer, breast cancer, and cutaneous melanoma. In primary brain tumors, such as gliomas, recent advances in DNA methylation profiling have allowed for a comprehensive molecular classification. Such data provide prognostic information, in addition to helping predict patient response to specific systemic therapies. However, epigenetic alterations of metastatic brain tumors with specific biological and translational relevance still require much further exploration. Using the widely employed Illumina Infinium HumanMethylation 450K platform, we have generated a cohort of genome-wide DNA methylomes from ninety-six needle-dissected BM specimens from patients with lung cancer, breast cancer, and cutaneous melanoma with clinical, pathological, and demographic annotations. This resource offers an unprecedented and unique opportunity to identify novel DNA methylation features influencing the behavior of brain metastasis, and thus accelerate the discovery of BM-specific theranostic epigenetic alterations.


Assuntos
Neoplasias Encefálicas , Metilação de DNA , Epigenômica , Adulto , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/secundário , DNA de Neoplasias , Feminino , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica/genética , Metástase Neoplásica/fisiopatologia , Análise de Sequência de DNA
7.
Int J Infect Dis ; 77: 18-22, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30243910

RESUMO

We report the case of a 69-year-old female who presented with a chronic nasal skin rash, new onset focal seizure, and a cerebral ring-enhancing lesion after a year of improper nasal irrigation. Despite aggressive and novel anti-amoebic treatment, she died as a result of a Balamuthia mandrillaris brain infection.


Assuntos
Balamuthia mandrillaris/isolamento & purificação , Encefalopatias/diagnóstico por imagem , Encéfalo/parasitologia , Lavagem Nasal/efeitos adversos , Idoso , Encéfalo/diagnóstico por imagem , Encefalopatias/etiologia , Encefalopatias/parasitologia , Exantema/tratamento farmacológico , Exantema/parasitologia , Evolução Fatal , Feminino , Humanos , Nariz/efeitos dos fármacos , Nariz/parasitologia , Sulfametoxazol/uso terapêutico , Trimetoprima/uso terapêutico
8.
J Transl Med ; 16(1): 179, 2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-29958537

RESUMO

Following publication of the original article [1], the authors reported an error in the spelling of one of the author names. In this Correction the incorrect and correct author names are indicated and the author name has been updated in the original publication. The authors also reported an error in the Methods section of the original article. In this Correction the incorrect and correct versions of the affected sentence are indicated. The original article has not been updated with regards to the error in the Methods section.

9.
J Transl Med ; 16(1): 142, 2018 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-29843811

RESUMO

BACKGROUND: Standard therapy for glioblastoma includes surgery, radiotherapy, and temozolomide. This Phase 3 trial evaluates the addition of an autologous tumor lysate-pulsed dendritic cell vaccine (DCVax®-L) to standard therapy for newly diagnosed glioblastoma. METHODS: After surgery and chemoradiotherapy, patients were randomized (2:1) to receive temozolomide plus DCVax-L (n = 232) or temozolomide and placebo (n = 99). Following recurrence, all patients were allowed to receive DCVax-L, without unblinding. The primary endpoint was progression free survival (PFS); the secondary endpoint was overall survival (OS). RESULTS: For the intent-to-treat (ITT) population (n = 331), median OS (mOS) was 23.1 months from surgery. Because of the cross-over trial design, nearly 90% of the ITT population received DCVax-L. For patients with methylated MGMT (n = 131), mOS was 34.7 months from surgery, with a 3-year survival of 46.4%. As of this analysis, 223 patients are ≥ 30 months past their surgery date; 67 of these (30.0%) have lived ≥ 30 months and have a Kaplan-Meier (KM)-derived mOS of 46.5 months. 182 patients are ≥ 36 months past surgery; 44 of these (24.2%) have lived ≥ 36 months and have a KM-derived mOS of 88.2 months. A population of extended survivors (n = 100) with mOS of 40.5 months, not explained by known prognostic factors, will be analyzed further. Only 2.1% of ITT patients (n = 7) had a grade 3 or 4 adverse event that was deemed at least possibly related to the vaccine. Overall adverse events with DCVax were comparable to standard therapy alone. CONCLUSIONS: Addition of DCVax-L to standard therapy is feasible and safe in glioblastoma patients, and may extend survival. Trial registration Funded by Northwest Biotherapeutics; Clinicaltrials.gov number: NCT00045968; https://clinicaltrials.gov/ct2/show/NCT00045968?term=NCT00045968&rank=1 ; initially registered 19 September 2002.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/terapia , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Glioblastoma/imunologia , Glioblastoma/terapia , Adulto , Idoso , Neoplasias Encefálicas/diagnóstico , Vacinas Anticâncer/efeitos adversos , Determinação de Ponto Final , Feminino , Glioblastoma/diagnóstico , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Análise de Sobrevida , Resultado do Tratamento , Adulto Jovem
10.
J Neurooncol ; 136(1): 181-188, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29098571

RESUMO

Appropriate management of adult gliomas requires an accurate histopathological diagnosis. However, the heterogeneity of gliomas can lead to misdiagnosis and undergrading, especially with biopsy. We evaluated the role of preoperative relative cerebral blood volume (rCBV) analysis in conjunction with histopathological analysis as a predictor of overall survival and risk of undergrading. We retrospectively identified 146 patients with newly diagnosed gliomas (WHO grade II-IV) that had undergone preoperative MRI with rCBV analysis. We compared overall survival by histopathologically determined WHO tumor grade and by rCBV using Kaplan-Meier survival curves and the Cox proportional hazards model. We also compared preoperative imaging findings and initial histopathological diagnosis in 13 patients who underwent biopsy followed by subsequent resection. Survival curves by WHO grade and rCBV tier similarly separated patients into low, intermediate, and high-risk groups with shorter survival corresponding to higher grade or rCBV tier. The hazard ratio for WHO grade III versus II was 3.91 (p = 0.018) and for grade IV versus II was 11.26 (p < 0.0001) and the hazard ratio for each increase in 1.0 rCBV units was 1.12 (p < 0.002). Additionally, 3 of 13 (23%) patients initially diagnosed by biopsy were upgraded on subsequent resection. Preoperative rCBV was elevated at least one standard deviation above the mean in the 3 upgraded patients, suggestive of undergrading, but not in the ten concordant diagnoses. In conclusion, rCBV can predict overall survival similarly to pathologically determined WHO grade in patients with gliomas. Discordant rCBV analysis and histopathology may help identify patients at higher risk for undergrading.


Assuntos
Neoplasias Encefálicas/irrigação sanguínea , Volume Sanguíneo Cerebral , Glioma/irrigação sanguínea , Adulto , Idoso , Biópsia , Determinação do Volume Sanguíneo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Feminino , Glioma/diagnóstico , Glioma/patologia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Período Pré-Operatório , Fatores de Risco
11.
Biochim Biophys Acta Rev Cancer ; 1868(1): 273-276, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28554666

RESUMO

The presence of human cytomegalovirus (HCMV) and glioblastoma multiforme (GBM), first established in 2002, has developed into an area of considerable interest and controversy. Numerous studies have found evidence of possible HCMV infection of GBM tumor cells as well as myriad onco- and immunomodulatory properties exhibited by HCMV antigens and transcripts, while recent reports have failed to detect HCMV particles in GBM and question the virus' role in tumor progression. This review highlights the known immunomodulatory properties of HCMV, independent of GBM infection status, that help drive the virus from peripheral blood into the vital tissues and subsequently dampen local immune response, assisting GBM tumors in evading immune surveillance and contributing to the disease's poor prognosis. Emerging antiviral approaches to treating GBM, including antiviral drugs and immunotherapies directed against HCMV, are also examined.


Assuntos
Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Citomegalovirus/imunologia , Glioblastoma/imunologia , Glioblastoma/patologia , Imunomodulação/imunologia , Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Progressão da Doença , Glioblastoma/virologia , Humanos
12.
Cell Syst ; 4(5): 516-529.e7, 2017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28365151

RESUMO

We present a systems strategy that facilitated the development of a molecular signature for glioblastoma (GBM), composed of 33 cell-surface transmembrane proteins. This molecular signature, GBMSig, was developed through the integration of cell-surface proteomics and transcriptomics from patient tumors in the REMBRANDT (n = 228) and TCGA datasets (n = 547) and can separate GBM patients from control individuals with a Matthew's correlation coefficient value of 0.87 in a lock-down test. Functionally, 17/33 GBMSig proteins are associated with transforming growth factor ß signaling pathways, including CD47, SLC16A1, HMOX1, and MRC2. Knockdown of these genes impaired GBM invasion, reflecting their role in disease-perturbed changes in GBM. ELISA assays for a subset of GBMSig (CD44, VCAM1, HMOX1, and BIGH3) on 84 plasma specimens from multiple clinical sites revealed a high degree of separation of GBM patients from healthy control individuals (area under the curve is 0.98 in receiver operating characteristic). In addition, a classifier based on these four proteins differentiated the blood of pre- and post-tumor resections, demonstrating potential clinical value as biomarkers.


Assuntos
Perfilação da Expressão Gênica/métodos , Glioblastoma/metabolismo , Proteínas de Membrana/metabolismo , Biomarcadores Tumorais , Neoplasias Encefálicas/genética , Diferenciação Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Biologia Computacional/métodos , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/genética , Humanos , Proteínas de Membrana/genética , Proteômica/métodos , Biologia de Sistemas/métodos , Transcriptoma/genética , Fator de Crescimento Transformador beta/metabolismo
13.
Oncotarget ; 8(16): 25989-25999, 2017 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-27517625

RESUMO

Glioblastoma multiforme (GBM) is a rapidly progressive brain tumor with a median survival of 15-19 months. Therapeutic resistance and recurrence of the disease is attributed to cancer stem cells (CSC). Here, we report that CMV70-3P miRNA encoded by CMV increases GBM CSC stemness. Inhibition of CMV70-3P expression using oligo inhibitors significantly attenuated the ability of primary glioma cells to proliferate and form neurospheres. At the molecular level, we show that CM70-3P increases expression of cellular SOX2. Collectively, these findings indicate that CMV70-3P is a potential regulator of CMV- mediated glioma progression and cancer stemness.


Assuntos
Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/virologia , Citomegalovirus/fisiologia , Glioma/etiologia , Glioma/metabolismo , MicroRNAs , Células-Tronco Neoplásicas/metabolismo , RNA Viral , Antígeno AC133/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Expressão Gênica , Glioma/tratamento farmacológico , Glioma/patologia , Humanos , Fenótipo , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/genética
14.
Cancer Lett ; 384: 79-85, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27777041

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive neoplastic brain tumor in humans with a median survival of less than 2 years. It is therefore critical to understand the mechanism of glioma progression and to identify future targets for intervention. We investigate the mechanisms of cytomegalovirus as an oncomodulatory agent implicated in glioma progression, as well as immunosuppression. This review provides a comprehensive evaluation of recent investigative developments concerning the role of CMV in cellular processes during glioma growth. The manners in which CMV and its viral products interact with regulatory cellular signaling pathways in the host are of primary interest. Here, we examine some of the most significant oncomodulatory effects that CMV can confer in brain tumors, including the inhibition of apoptosis and promoting the growth of glioma stem cells, which are tightly linked to tumor survival and recurrence.


Assuntos
Neoplasias Encefálicas/virologia , Transformação Celular Viral , Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Glioma/virologia , Infecções Tumorais por Vírus/virologia , Animais , Apoptose , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Ciclo Celular , Proliferação de Células , Citomegalovirus/imunologia , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Progressão da Doença , Glioma/epidemiologia , Glioma/imunologia , Glioma/patologia , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Evasão Tumoral , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/metabolismo
15.
Stem Cells ; 34(9): 2276-89, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27354342

RESUMO

Glioblastoma multiforme (GBM) is the most common and lethal adult brain tumor. Resistance to standard radiation and chemotherapy is thought to involve survival of GBM cancer stem cells (CSCs). To date, no single marker for identifying GBM CSCs has been able to capture the diversity of CSC populations, justifying the needs for additional CSC markers for better characterization. Employing targeted mass spectrometry, here we present five cell-surface markers HMOX1, SLC16A1, CADM1, SCAMP3, and CLCC1 which were found to be elevated in CSCs relative to healthy neural stem cells (NSCs). Transcriptomic analyses of REMBRANDT and TCGA compendiums also indicated elevated expression of these markers in GBM relative to controls and non-GBM diseases. Two markers SLC16A1 and HMOX1 were found to be expressed among pseudopalisading cells that reside in the hypoxic region of GBM, substantiating the histopathological hallmarks of GBM. In a prospective study (N = 8) we confirmed the surface expression of HMOX1 on freshly isolated primary GBM cells (P0). Employing functional assays that are known to evaluate stemness, we demonstrate that elevated HMOX1 expression is associated with stemness in GBM and can be modulated through TGFß. siRNA-mediated silencing of HMOX1 impaired GBM invasion-a phenomenon related to poor prognosis. In addition, surgical resection of GBM tumors caused declines (18% ± 5.1SEM) in the level of plasma HMOX1 as measured by ELISA, in 8/10 GBM patients. These findings indicate that HMOX1 is a robust predictor of GBM CSC stemness and pathogenesis. Further understanding of the role of HMOX1 in GBM may uncover novel therapeutic approaches. Stem Cells 2016;34:2276-2289.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Heme Oxigenase-1/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fator de Crescimento Transformador beta/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Autorrenovação Celular , Glioblastoma/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Invasividade Neoplásica , Células-Tronco Neurais/metabolismo , Prognóstico , Esferoides Celulares/metabolismo , Simportadores/metabolismo
16.
Cancer Res ; 75(15): 3065-76, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26239477

RESUMO

Glioblastoma (GBM) is the most common and aggressive human brain tumor. Human cytomegalovirus (HCMV) immediate-early (IE) proteins that are endogenously expressed in GBM cells are strong viral transactivators with oncogenic properties. Here, we show how HCMV IEs are preferentially expressed in glioma stem-like cells (GSC), where they colocalize with the other GBM stemness markers, CD133, Nestin, and Sox2. In patient-derived GSCs that are endogenously infected with HCMV, attenuating IE expression by an RNAi-based strategy was sufficient to inhibit tumorsphere formation, Sox2 expression, cell-cycle progression, and cell survival. Conversely, HCMV infection of HMCV-negative GSCs elicited robust self-renewal and proliferation of cells that could be partially reversed by IE attenuation. In HCMV-positive GSCs, IE attenuation induced a molecular program characterized by enhanced expression of mesenchymal markers and proinflammatory cytokines, resembling the therapeutically resistant GBM phenotype. Mechanistically, HCMV/IE regulation of Sox2 occurred via inhibition of miR-145, a negative regulator of Sox2 protein expression. In a spontaneous mouse model of glioma, ectopic expression of the IE1 gene (UL123) specifically increased Sox2 and Nestin levels in the IE1-positive tumors, upregulating stemness and proliferation markers in vivo. Similarly, human GSCs infected with the HCMV strain Towne but not the IE1-deficient strain CR208 showed enhanced growth as tumorspheres and intracranial tumor xenografts, compared with mock-infected human GSCs. Overall, our findings offer new mechanistic insights into how HCMV/IE control stemness properties in GBM cells.


Assuntos
Antígenos Virais/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Glioblastoma/patologia , Glioblastoma/virologia , Proteínas Imediatamente Precoces/metabolismo , Animais , Antígenos Virais/genética , Apoptose/genética , Neoplasias Encefálicas/metabolismo , Citomegalovirus/genética , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/patologia , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Glioma/genética , Glioma/patologia , Humanos , Proteínas Imediatamente Precoces/genética , Camundongos Endogâmicos BALB C , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/virologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células Tumorais Cultivadas
17.
Oncotarget ; 6(6): 3977-87, 2015 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-25738357

RESUMO

Oncolytic gene therapy using viral vectors may provide an attractive therapeutic option for malignant gliomas. These viral vectors are designed in a way to selectively target tumor cells and spare healthy cells. To determine the translational impact, it is imperative to assess the factors that interfere with the anti-glioma effects of the oncolytic adenoviral vectors. In the current study, we evaluated the efficacy of survivin-driven oncolytic adenoviruses pseudotyping with adenoviral fiber knob belonging to the adenoviral serotype 3, 11 and 35 in their ability to kill glioblastoma (GBM) cells selectively without affecting normal cells. Our results indicate that all recombinant vectors used in the study can effectively target GBM in vitro with high specificity, especially the 3 knob-modified vector. Using intracranial U87 and U251 GBM xenograft models we have also demonstrated that treatment with Conditionally Replicative Adenovirus (CRAd-S-5/3) vectors can effectively regress tumor. However, in several patient-derived GBM cell lines, cells exhibited resistance to the CRAd infection as evident from the diminishing effects of autophagy. To improve therapeutic response, tumor cells were pretreated with tamoxifen. Our preliminary data suggest that tamoxifen sensitizes glioblastoma cells towards oncolytic treatment with CRAd-S-5/3, which may prove useful for GBM in future experimental therapy.


Assuntos
Adenoviridae/fisiologia , Antineoplásicos Hormonais/farmacologia , Neoplasias Encefálicas/terapia , Glioblastoma/terapia , Terapia Viral Oncolítica/métodos , Tamoxifeno/farmacologia , Adenoviridae/genética , Animais , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Linhagem Celular Tumoral , Terapia Combinada , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioblastoma/virologia , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Methods Mol Biol ; 1119: 165-96, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24639224

RESUMO

An increased awareness of the potential oncomodulatory properties of human cytomegalovirus (HCMV) has evolved over the last decade. We first reported the presence of HCMV in human glioblastomas, and subsequently these findings have been corroborated by other groups. However, some controversy has been associated with the immunohistochemical and in situ hybridization techniques used, since standard immunohistochemical and in situ hybridization techniques have been insufficient to detect low level HCMV antigens and nucleic acids in some tumor tissues. Here, we present detailed methods that can be used for the sensitive detection of low level HCMV antigens and nucleic acids in human glioblastoma specimens. Using these techniques, HCMV is frequently detected in frozen and formalin fixed paraffin-embedded tissue specimens. Furthermore, we demonstrate how human primary glioblastoma cells can be cultured in vitro, and how these cells can be used for detection of HCMV by immunofluorescence, in situ hybridization, western blot, and RT-PCR.


Assuntos
Citomegalovirus/isolamento & purificação , DNA Viral/isolamento & purificação , Biologia Molecular/métodos , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Glioblastoma/virologia , Humanos , Hibridização In Situ
20.
Curr Opin Oncol ; 25(6): 682-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24097102

RESUMO

PURPOSE OF REVIEW: First described in 2002, the presence and role of human cytomegalovirus (HCMV) infection in glioblastoma (GBM) has remained a controversial topic. New research indicates HCMV gene products likely promote GBM pathogenesis and that therapies aimed at HCMV might influence disease progression. RECENT FINDINGS: Recently, investigators have begun to analyze HCMV genome and proteins present in GBM cells in vivo. Furthermore, the research has demonstrated that several HCMV gene products that have oncomodulatory properties are expressed in GBM and may be impacting tumor pathogenesis in vivo. These HCMV gene products modulate GBM proliferation, apoptosis, angiogenesis, invasion and immune evasion. A recent mouse model provides mechanistic information as to how CMV may promote gliomagenesis in the setting of tumor suppressor dysfunction and STAT3 signaling. In addition, clinical outcomes of GBM patients are associated with the degree of HCMV infection. Novel therapies aimed at direct antiviral and immunotherapy approaches to HCMV suggest that these modalities may impact the future treatment of this disease. SUMMARY: A more precise understanding of the role of HCMV infection in gliomagenesis and GBM pathogenesis could reveal novel therapeutic and preventive strategies.


Assuntos
Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/virologia , Infecções por Citomegalovirus/complicações , Citomegalovirus/isolamento & purificação , Glioblastoma/epidemiologia , Glioblastoma/virologia , Infecções Tumorais por Vírus/complicações , Animais , Antivirais/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Infecções por Citomegalovirus/epidemiologia , Feminino , Ganciclovir/análogos & derivados , Ganciclovir/uso terapêutico , Regulação Neoplásica da Expressão Gênica/imunologia , Regulação Viral da Expressão Gênica/imunologia , Glioblastoma/tratamento farmacológico , Humanos , Masculino , Camundongos , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Infecções Tumorais por Vírus/epidemiologia , Valganciclovir
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...