Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
JAMA Oncol ; 9(1): 112-121, 2023 01 01.
Article in English | MEDLINE | ID: mdl-36394838

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Temozolomide/therapeutic use , Prospective Studies , Brain Neoplasms/pathology , Recurrence , Dendritic Cells/pathology , Vaccination
2.
iScience ; 25(6): 104395, 2022 Jun 17.
Article in English | MEDLINE | ID: mdl-35637733

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-30855281

ABSTRACT

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.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus/metabolism , Glioblastoma , Neoplasms, Experimental , Neovascularization, Pathologic , Pericytes , Animals , Cell Line, Tumor , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/pathology , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/virology , Humans , Lymphokines/metabolism , Mice , NIH 3T3 Cells , Neoplasm Proteins/metabolism , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/virology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/virology , Pericytes/metabolism , Pericytes/pathology , Platelet-Derived Growth Factor/metabolism
4.
Neurosurgery ; 84(3): E168-E170, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30629225

ABSTRACT

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.


Subject(s)
Brain Neoplasms/radiotherapy , Neurosurgeons/standards , Practice Guidelines as Topic/standards , Radiosurgery/standards , Adult , Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Congresses as Topic/standards , Disease Progression , Female , Humans , Male , Quality of Life , Radiosurgery/mortality , Tumor Burden
5.
Sci Data ; 5: 180245, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30398472

ABSTRACT

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.


Subject(s)
Brain Neoplasms , DNA Methylation , Epigenomics , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Brain Neoplasms/secondary , DNA, Neoplasm , Female , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Middle Aged , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology , Sequence Analysis, DNA
6.
Nat Commun ; 9(1): 4627, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30401823

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Neoplasm Metastasis/genetics , Algorithms , Brain Neoplasms/pathology , DNA, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Melanoma , Skin Neoplasms , Supervised Machine Learning , Melanoma, Cutaneous Malignant
7.
Int J Infect Dis ; 77: 18-22, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30243910

ABSTRACT

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.


Subject(s)
Balamuthia mandrillaris/isolation & purification , Brain Diseases/diagnostic imaging , Brain/parasitology , Nasal Lavage/adverse effects , Aged , Brain/diagnostic imaging , Brain Diseases/etiology , Brain Diseases/parasitology , Exanthema/drug therapy , Exanthema/parasitology , Fatal Outcome , Female , Humans , Nose/drug effects , Nose/parasitology , Sulfamethoxazole/therapeutic use , Trimethoprim/therapeutic use
8.
J Transl Med ; 16(1): 179, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29958537

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-29843811

ABSTRACT

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.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cancer Vaccines/immunology , Dendritic Cells/immunology , Glioblastoma/immunology , Glioblastoma/therapy , Adult , Aged , Brain Neoplasms/diagnosis , Cancer Vaccines/adverse effects , Endpoint Determination , Female , Glioblastoma/diagnosis , Humans , Male , Middle Aged , Prognosis , Survival Analysis , Treatment Outcome , Young Adult
10.
J Neurooncol ; 136(1): 181-188, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29098571

ABSTRACT

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.


Subject(s)
Brain Neoplasms/blood supply , Cerebral Blood Volume , Glioma/blood supply , Adult , Aged , Biopsy , Blood Volume Determination , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Female , Glioma/diagnosis , Glioma/pathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Preoperative Period , Risk Factors
11.
Biochim Biophys Acta Rev Cancer ; 1868(1): 273-276, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28554666

ABSTRACT

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.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cytomegalovirus/immunology , Glioblastoma/immunology , Glioblastoma/pathology , Immunomodulation/immunology , Brain Neoplasms/virology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/pathology , Cytomegalovirus Infections/virology , Disease Progression , Glioblastoma/virology , Humans
12.
Cell Syst ; 4(5): 516-529.e7, 2017 05 24.
Article in English | MEDLINE | ID: mdl-28365151

ABSTRACT

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.


Subject(s)
Gene Expression Profiling/methods , Glioblastoma/metabolism , Membrane Proteins/metabolism , Biomarkers, Tumor , Brain Neoplasms/genetics , Cell Differentiation , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Computational Biology/methods , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Humans , Membrane Proteins/genetics , Proteomics/methods , Systems Biology/methods , Transcriptome/genetics , Transforming Growth Factor beta/metabolism
13.
Oncotarget ; 8(16): 25989-25999, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-27517625

ABSTRACT

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.


Subject(s)
Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Glioma/etiology , Glioma/metabolism , MicroRNAs , Neoplastic Stem Cells/metabolism , RNA, Viral , AC133 Antigen/metabolism , Cell Line, Tumor , Cell Movement/genetics , Gene Expression , Glioma/drug therapy , Glioma/pathology , Humans , Phenotype , Promoter Regions, Genetic , SOXB1 Transcription Factors/genetics
14.
Cancer Lett ; 384: 79-85, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27777041

ABSTRACT

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.


Subject(s)
Brain Neoplasms/virology , Cell Transformation, Viral , Cytomegalovirus Infections/virology , Cytomegalovirus/pathogenicity , Glioma/virology , Tumor Virus Infections/virology , Animals , Apoptosis , Brain Neoplasms/epidemiology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Cycle , Cell Proliferation , Cytomegalovirus/immunology , Cytomegalovirus/metabolism , Cytomegalovirus Infections/epidemiology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/metabolism , Disease Progression , Glioma/epidemiology , Glioma/immunology , Glioma/pathology , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Tumor Escape , Tumor Virus Infections/epidemiology , Tumor Virus Infections/immunology , Tumor Virus Infections/metabolism
15.
Stem Cells ; 34(9): 2276-89, 2016 09.
Article in English | MEDLINE | ID: mdl-27354342

ABSTRACT

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.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Heme Oxygenase-1/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Transforming Growth Factor beta/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cell Self Renewal , Glioblastoma/metabolism , Humans , Membrane Proteins/metabolism , Monocarboxylic Acid Transporters/metabolism , Neoplasm Invasiveness , Neural Stem Cells/metabolism , Prognosis , Spheroids, Cellular/metabolism , Symporters/metabolism
16.
Cancer Res ; 75(15): 3065-76, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26239477

ABSTRACT

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.


Subject(s)
Antigens, Viral/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/virology , Glioblastoma/pathology , Glioblastoma/virology , Immediate-Early Proteins/metabolism , Animals , Antigens, Viral/genetics , Apoptosis/genetics , Brain Neoplasms/metabolism , Cytomegalovirus/genetics , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/pathology , Disease Models, Animal , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioma/genetics , Glioma/pathology , Humans , Immediate-Early Proteins/genetics , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/virology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Tumor Cells, Cultured
17.
Oncotarget ; 6(6): 3977-87, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25738357

ABSTRACT

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.


Subject(s)
Adenoviridae/physiology , Antineoplastic Agents, Hormonal/pharmacology , Brain Neoplasms/therapy , Glioblastoma/therapy , Oncolytic Virotherapy/methods , Tamoxifen/pharmacology , Adenoviridae/genetics , Animals , Autophagy/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/virology , Cell Line, Tumor , Combined Modality Therapy , Female , Glioblastoma/genetics , Glioblastoma/pathology , Glioblastoma/virology , Humans , Mice , Xenograft Model Antitumor Assays
18.
Methods Mol Biol ; 1119: 165-96, 2014.
Article in English | MEDLINE | ID: mdl-24639224

ABSTRACT

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.


Subject(s)
Cytomegalovirus/isolation & purification , DNA, Viral/isolation & purification , Molecular Biology/methods , Cytomegalovirus/pathogenicity , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/virology , Glioblastoma/virology , Humans , In Situ Hybridization
20.
Clin Cancer Res ; 19(23): 6473-83, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24170543

ABSTRACT

PURPOSE: Cidofovir (CDV) is an U.S. Food and Drug Administration (FDA)-approved nucleoside antiviral agent used to treat severe human cytomegalovirus (HCMV) infection. Until now, no clear therapeutic effects of CDV have been reported outside of the setting of viral infection, including a potential role for CDV as an antineoplastic agent for the treatment of brain tumors. EXPERIMENTAL DESIGN: We investigated the cytotoxicity of CDV against the glioblastoma cells, U87MG and primary SF7796, both in vitro and in vivo, using an intracranial xenograft model. Standard techniques for cell culturing, immunohistochemistry, Western blotting, and real-time PCR were employed. The survival of athymic mice (n = 8-10 per group) bearing glioblastoma tumors, treated with CDV alone or in combination with radiation, was analyzed by the Kaplan-Meier method and evaluated with a two-sided log-rank test. RESULTS: CDV possesses potent antineoplastic activity against HCMV-infected glioblastoma cells. This activity is associated with the inhibition of HCMV gene expression and with activation of cellular apoptosis. Surprisingly, we also determined that CDV induces glioblastoma cell death in the absence of HCMV infection. CDV is incorporated into tumor cell DNA, which promotes double-stranded DNA breaks and induces apoptosis. In the setting of ionizing radiotherapy, the standard of care for glioblastoma in humans, CDV augments radiation-induced DNA damage and, further, promotes tumor cell death. Combination therapy with CDV and radiotherapy significantly extended the survival of mice bearing intracranial glioblastoma tumors. CONCLUSION: We have identified a novel antiglioma property of the FDA-approved drug CDV, which heightens the cytotoxic effect of radiotherapy, the standard of care therapy for glioblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Cytosine/analogs & derivatives , Glioblastoma/drug therapy , Organophosphonates/pharmacology , Animals , Antineoplastic Agents/metabolism , Apoptosis , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cidofovir , Cytosine/metabolism , Cytosine/pharmacology , DNA Breaks, Double-Stranded , DNA Replication , Female , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Organophosphonates/metabolism , Spheroids, Cellular/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL