Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38413483

ABSTRACT

BACKGROUND: Following catheter ablation, vascular access management involves potential complications and prolonged recovery. Recently, suture-mediated closure (SMC) devices were approved for venous access procedures. The objective of this study is to evaluate the safety of a commercially available SMC for multiple access site venous closure by duplex ultrasound (DUS) in asymptomatic subjects with non-visible complications. METHODS: Thirty-six subjects (63 ± 10.7 years old, 12 female) were enrolled. Following catheter ablation for atrial fibrillation, all subjects had SMC of every venous access site. Subjects underwent DUS of femoral veins and arteries. DUS was performed at discharge, and again at 30 days. Subjects were evaluated for clinically apparent vascular complications. RESULTS: Mean procedure duration was 138.6 min, and the time to hemostasis was 3.1 min/access site and 9.5 min/subject. Median time to ambulation was 193.5 min, and median time to discharge was 5.95 h, with discharge as early as 2.4 h. A median of 2 sheaths/vein and a median of 2 SMC devices/vein were used. There were no major complications and a 16.7% (6/36) minor complication rate at discharge. All complications resolved at 30 days. The complication rate was not higher in patients with 2 SMC per access site as compared to the patients who just received 1 SMC per access site. CONCLUSIONS: This study demonstrates the safety of multi-access closure using SMC, following catheter ablation procedures, for closure of sites that use sheath sizes from ≤ 8F to ≥ 15F and for those that use 2 or more SMCs per access site.

2.
Future Sci OA ; 9(6): FSO864, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37228857

ABSTRACT

Traumatic brain injury (TBI) is a significant cause of mortality and morbidity worldwide resulting from falls, car accidents, sports, and blast injuries. TBI is characterized by severe, life-threatening consequences due to neuroinflammation in the brain. Contact and collision sports lead to higher disability and death rates among young adults. Unfortunately, no therapy or drug protocol currently addresses the complex pathophysiology of TBI, leading to the long-term chronic neuroinflammatory assaults. However, the immune response plays a crucial role in tissue-level injury repair. This review aims to provide a better understanding of TBI's immunobiology and management protocols from an immunopathological perspective. It further elaborates on the risk factors, disease outcomes, and preclinical studies to design precisely targeted interventions for enhancing TBI outcomes.


Traumatic brain injury (TBI) is a leading cause of death and disability worldwide due to falls, car accidents, sports and blast injuries. TBI causes severe, life-threatening consequences due to inflammation in the brain. Unfortunately, no current therapy or drug protocol can address the complexity of TBI, leading to long-term chronic inflammation. However, the immune response plays a crucial role in repairing injured brain tissue. This review aims to provide a better understanding of TBI's immunobiology and management protocols to design targeted interventions for better outcomes in TBI patients.

3.
Future Sci OA ; 9(4): FSO851, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37090492

ABSTRACT

The cerebrospinal fluid (CSF) is a clear ultrafiltrate of blood that envelopes and protects the central nervous system while regulating neuronal function through the maintenance of interstitial fluid homeostasis in the brain. Due to its anatomic location and physiological functions, the CSF can provide a reliable source of biomarkers for the diagnosis and treatment monitoring of different neurological diseases, including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and primary and secondary brain malignancies. The incorporation of CSF biomarkers into the drug discovery and development can improve the efficiency of drug development and increase the chances of success. This review aims to consolidate the current use of CSF biomarkers in clinical practice and explore future perspectives for the field.


Cerebrospinal fluid (CSF) is a clear fluid that protects our brain and spinal cord, and can help diagnose and monitor neurological diseases like Alzheimer's and Parkinson's. Biomarkers in CSF are like clues that help doctors and researchers better understand these diseases. By using CSF biomarkers, doctors can diagnose and monitor patients more accurately, while researchers can develop more effective treatments. This review looks at how we use CSF biomarkers in medicine and how they might help us in the future. Better understanding of CSF biomarkers can improve the lives of people living with neurological diseases.

4.
J Transl Med ; 20(1): 620, 2022 12 26.
Article in English | MEDLINE | ID: mdl-36572880

ABSTRACT

Glioblastoma is the most lethal form of brain tumor with a recurrence rate of almost 90% and a survival time of only 15 months post-diagnosis. It is a highly heterogeneous, aggressive, and extensively studied tumor. Multiple studies have proposed therapeutic approaches to mitigate or improve the survival for patients with glioblastoma. In this article, we review the loss of the 5'-methylthioadenosine phosphorylase (MTAP) gene as a potential therapeutic approach for treating glioblastoma. MTAP encodes a metabolic enzyme required for the metabolism of polyamines and purines leading to DNA synthesis. Multiple studies have explored the loss of this gene and have shown its relevance as a therapeutic approach to glioblastoma tumor mitigation; however, other studies show that the loss of MTAP does not have a major impact on the course of the disease. This article reviews the contrasting findings of MTAP loss with regard to mitigating the effects of glioblastoma, and also focuses on multiple aspects of MTAP loss in glioblastoma by providing insights into the known findings and some of the unexplored areas of this field where new approaches can be imagined for novel glioblastoma therapeutics.


Subject(s)
Glioblastoma , Humans , Glioblastoma/genetics , Glioblastoma/therapy , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism
5.
Sensors (Basel) ; 22(3)2022 Jan 19.
Article in English | MEDLINE | ID: mdl-35161502

ABSTRACT

Wearable devices use sensors to evaluate physiological parameters, such as the heart rate, pulse rate, number of steps taken, body fat and diet. The continuous monitoring of physiological parameters offers a potential solution to assess personal healthcare. Identifying outliers or anomalies in heart rates and other features can help identify patterns that can play a significant role in understanding the underlying cause of disease states. Since anomalies are present within the vast amount of data generated by wearable device sensors, identifying anomalies requires accurate automated techniques. Given the clinical significance of anomalies and their impact on diagnosis and treatment, a wide range of detection methods have been proposed to detect anomalies. Much of what is reported herein is based on previously published literature. Clinical studies employing wearable devices are also increasing. In this article, we review the nature of the wearables-associated data and the downstream processing methods for detecting anomalies. In addition, we also review supervised and un-supervised techniques as well as semi-supervised methods that overcome the challenges of missing and un-annotated healthcare data.


Subject(s)
Data Analysis , Wearable Electronic Devices , Algorithms , Heart Rate
6.
World J Stem Cells ; 12(10): 1067-1079, 2020 Oct 26.
Article in English | MEDLINE | ID: mdl-33178392

ABSTRACT

Coronavirus disease 2019 (COVID-19), a pandemic disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), is growing at an exponential rate worldwide. Manifestations of this disease are heterogeneous; however, advanced cases often exhibit various acute respiratory distress syndrome-like symptoms, systemic inflammatory reactions, coagulopathy, and organ involvements. A common theme in advanced COVID-19 is unrestrained immune activation, classically referred to as a "cytokine storm", as well as deficiencies in immune regulatory mechanisms such as T regulatory cells. While mesenchymal stem cells (MSCs) themselves are objects of cytokine regulation, they can secrete cytokines to modulate immune cells by inducing anti-inflammatory regulatory Treg cells, macrophages and neutrophils; and by reducing the activation of T and B cells, dendritic and nature killer cells. Consequently, they have therapeutic potential for treating severe cases of COVID-19. Here we discuss the unique ability of MSCs, to act as a "living anti-inflammatory", which can "rebalance" the cytokine/immune responses to restore equilibrium. We also discuss current MSC trials and present different concepts for optimization of MSC therapy in patients with COVID-19 acute respiratory distress syndrome.

7.
CNS Oncol ; 8(2): CNS34, 2019 06.
Article in English | MEDLINE | ID: mdl-30855176

ABSTRACT

Aim: Genomically matched trials in primary brain tumors (PBTs) require recent tumor sequencing. We evaluated whether circulating tumor DNA (ctDNA) could facilitate genomic interrogation in these patients. Methods: Data from 419 PBT patients tested clinically with a ctDNA NGS panel at a CLIA-certified laboratory were analyzed. Results: A total of 211 patients (50%) had ≥1 somatic alteration detected. Detection was highest in meningioma (59%) and gliobastoma (55%). Single nucleotide variants were detected in 61 genes, with amplifications detected in ERBB2, MET, EGFR and others. Conclusion: Contrary to previous studies with very low yields, we found half of PBT patients had detectable ctDNA with genomically targetable off-label or clinical trial options for almost 50%. For those PBT patients with detectable ctDNA, plasma cfDNA genomic analysis is a clinically viable option for identifying genomically driven therapy options.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Circulating Tumor DNA/genetics , Glioblastoma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Brain Neoplasms/blood , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Child , Child, Preschool , Circulating Tumor DNA/blood , Female , Glioblastoma/blood , Glioblastoma/diagnosis , Glioblastoma/pathology , Humans , Male , Middle Aged , Mutation , Prognosis , Sequence Analysis, DNA , Young Adult
8.
Colloids Surf B Biointerfaces ; 161: 200-209, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29080504

ABSTRACT

Circulating tumor cells (CTCs) are extremely rare cells found in blood of metastatic cancer patients. There is a need for inexpensive technologies for fast enrichment of CTCs from large blood volumes. Previous data showed that antibody-conjugated lipid shell immuno-microbubbles (MBs) bind and isolate cells from biological fluids by flotation. Here, blood-stable MBs targeted to several surface markers for isolation of breast tumor cells were developed. MBs coated with anti-human EpCAM antibodies showed efficient binding of EpCAM+ breast cancer cell lines SKBR-3, MCF-7, and MDA-MB-453, whereas anti-human EGFR MBs showed binding of EpCAMLOW/NEGATIVE cell lines MDA-MB-231 and BT-549. Multitargeted anti-human EpCAM/EGFR MBs bound all cell lines with over 95% efficiency. Highly concentrated MB-bound tumor cells were collected in a microliter volume via an inverted vacuum-assisted harvesting setup. Using anti-EpCAM and/or anti-EpCAM/EGFR MBs, an efficient (70-90%) recovery and fast (30min) isolation of the above-mentioned cells and cell clusters was achieved from 7.5mL of spiked human blood. Using anti-EpCAM MBs and anti-EpCAM/EGFR MBs, cytokeratin-positive, CD45-negative CTCs were detected in 62.5% (10/16) of patients with metastatic breast cancer and CTC clusters were detected in 41.7% (5/12) of CTC-positive samples. Moreover, in some samples MBs isolated cytokeratin positive, CD45 negative tumor-derived microparticles. None of these structures were detected in blood from non-epithelial malignancies. The fast and inexpensive multitargeted platform for batch isolation of CTCs can promote research and clinical applications involving primary tumors and metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/blood , Cell Separation/methods , Microbubbles , Neoplastic Cells, Circulating/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Epithelial Cell Adhesion Molecule/metabolism , Female , Humans , Keratins/metabolism , MCF-7 Cells , Neoplastic Cells, Circulating/pathology
9.
Hum Antibodies ; 26(2): 95-101, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29036806

ABSTRACT

Immunotherapy is now at the forefront of cancer therapeutic development. Gliomas are a particularly aggressive form of brain cancer for which immunotherapy may hold promise. Pritumumab (also known in the literature as CLNH11, CLN-IgG, and ACA-11) was the first monoclonal antibody tested in cancer patients. Pritumumab is a natural human monoclonal antibody developed from a B lymphocyte isolated from a regional draining lymph node of a patient with cervical carcinoma. The antibody binds ecto-domain vimentin on the surface of cancer cells. Pritumumab was originally tested in clinical trials with brain cancer patients in Japan where it demonstrated therapeutic benefit. It was reported to be a safe and effective therapy for brain cancer patients at doses 5-10 fold less than currently approved antibodies. Phase I dose escalation clinical trials are now being planned with pritumumab for the near future. Here we review data on the development and characterization of pritumumab, and review clinical trails data assessing immunotherapeutic effects of pritumumab for glioma patients.


Subject(s)
Antibodies, Monoclonal/isolation & purification , Antineoplastic Agents, Immunological/isolation & purification , Brain Neoplasms/drug therapy , Glioma/drug therapy , Immunoglobulin G/isolation & purification , Vimentin/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Antineoplastic Agents, Immunological/metabolism , Antineoplastic Agents, Immunological/therapeutic use , B-Lymphocytes/chemistry , B-Lymphocytes/immunology , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Clinical Trials as Topic , Gene Expression , Glioma/genetics , Glioma/immunology , Glioma/mortality , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/therapeutic use , Immunotherapy/methods , Mice , Survival Analysis , Vimentin/antagonists & inhibitors , Vimentin/genetics , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 23(16): 4716-4723, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28420725

ABSTRACT

Purpose: Noninvasive drug biomarkers for the early assessment of tumor response can enable adaptive therapeutic decision-making and proof-of-concept studies for investigational drugs. Circulating tumor DNA (ctDNA) is released into the circulation by tumor cell turnover and has been shown to be detectable in urine.Experimental Design: We tested the hypothesis that dynamic changes in EGFR activating (exon 19del and L858R) and resistance (T790M) mutation levels detected in urine could inform tumor response within days of therapy for advanced non-small cell lung cancer (NSCLC) patients receiving osimertinib, a second-line third-generation anti-EGFR tyrosine kinase inhibitor.Results: Eight of nine evaluable NSCLC patients had detectable T790M-mutant DNA fragments in pretreatment baseline samples. Daily monitoring of mutations in urine indicated a pattern of intermittent spikes throughout week 1, suggesting apoptosis with an overall decrease in fragment numbers from baselines to day 7 preceding radiographic response assessed at 6 to 12 weeks.Conclusions: These findings suggest drug-induced tumor apoptosis within days of initial dosing. Daily sampling of ctDNA may enable early assessment of patient response and proof-of-concept studies for drug development. The modeling of tumor lysis through the day-to-day kinetics of ctDNA released into the blood and then into the urine is demonstrated in this proof-of-concept study in lung cancer patients receiving anti-EGFR tyrosine kinase inhibitors. This strategy may determine the specific clonal populations of cells which undergo apoptosis within the first week of therapy. This has important implications for developing combinational strategies to address inter- and intralesional heterogeneity and characterizing residual disease after initial drug exposure. Clin Cancer Res; 23(16); 4716-23. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Circulating Tumor DNA/urine , DNA, Neoplasm/urine , Lung Neoplasms/drug therapy , Piperazines/therapeutic use , Acrylamides , Aged , Aniline Compounds , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/urine , Circulating Tumor DNA/genetics , DNA, Neoplasm/genetics , Drug Monitoring , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Exons/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/urine , Middle Aged , Molecular Targeted Therapy , Mutation , Protein Kinase Inhibitors/therapeutic use , Time Factors , Treatment Outcome
11.
Clin Cancer Res ; 23(14): 3657-3666, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28096270

ABSTRACT

Purpose: Tumor-derived cell-free DNA (cfDNA) from urine of patients with cancer offers noninvasive biological material for detection of cancer-related molecular abnormalities such as mutations in Exon 2 of KRASExperimental Design: A quantitative, mutation-enrichment next-generation sequencing test for detecting KRASG12/G13 mutations in urine cfDNA was developed, and results were compared with clinical testing of archival tumor tissue and plasma cfDNA from patients with advanced cancer.Results: With 90 to 110 mL of urine, the KRASG12/G13 cfDNA test had an analytical sensitivity of 0.002% to 0.006% mutant copies in wild-type background. In 71 patients, the concordance between urine cfDNA and tumor was 73% (sensitivity, 63%; specificity, 96%) for all patients and 89% (sensitivity, 80%; specificity, 100%) for patients with urine samples of 90 to 110 mL. Patients had significantly fewer KRASG12/G13 copies in urine cfDNA during systemic therapy than at baseline or disease progression (P = 0.002). Compared with no changes or increases in urine cfDNA KRASG12/G13 copies during therapy, decreases in these measures were associated with longer median time to treatment failure (P = 0.03).Conclusions: A quantitative, mutation-enrichment next-generation sequencing test for detecting KRASG12/G13 mutations in urine cfDNA had good concordance with testing of archival tumor tissue. Changes in mutated urine cfDNA were associated with time to treatment failure. Clin Cancer Res; 23(14); 3657-66. ©2017 AACR.


Subject(s)
Biomarkers, Tumor/urine , Cell-Free Nucleic Acids/urine , Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/urine , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cell-Free Nucleic Acids/genetics , Female , Humans , Male , Middle Aged , Mutation , Neoplasms/pathology , Neoplasms/urine , Proto-Oncogene Proteins p21(ras)/genetics
12.
Oncotarget ; 8(14): 22370-22384, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-26517684

ABSTRACT

Transcription factors (TFs) are a major class of protein signaling molecules that play key cellular roles in cancers such as the highly lethal brain cancer-glioblastoma (GBM). However, the development of specific TF inhibitors has proved difficult owing to expansive protein-protein interfaces and the absence of hydrophobic pockets. We uniquely defined the dimerization surface as an expansive parental pharmacophore comprised of several regional daughter pharmacophores. We targeted the OLIG2 TF which is essential for GBM survival and growth, we hypothesized that small molecules able to fit each subpharmacophore would inhibit OLIG2 activation. The most active compound was OLIG2 selective, it entered the brain, and it exhibited potent anti-GBM activity in cell-based assays and in pre-clinical mouse orthotopic models. These data suggest that (1) our multiple pharmacophore approach warrants further investigation, and (2) our most potent compounds merit detailed pharmacodynamic, biophysical, and mechanistic characterization for potential preclinical development as GBM therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Brain Neoplasms/drug therapy , Drug Design , Glioblastoma/drug therapy , Guanidines/therapeutic use , Molecular Targeted Therapy , Nerve Tissue Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/chemistry , Cell Growth Processes , Cell Survival/genetics , Computer Simulation , Humans , Mice , Mice, Nude , Molecular Structure , Nerve Tissue Proteins/chemistry , Oligodendrocyte Transcription Factor 2 , Protein Binding , Protein Conformation , Small Molecule Libraries , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Sci Rep ; 6: 39479, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27995987

ABSTRACT

Sunlight has important biological effects in human skin. Ultraviolet (UV) light striking the epidermis catalyzes the synthesis of Vitamin D and triggers melanin production. Although a causative element in skin cancers, sunlight is also associated with positive health outcomes including reduced incidences of autoimmune diseases and cancers. The mechanisms, however, by which light affects immune function remain unclear. Here we describe direct photon sensing in human and mouse T lymphocytes, a cell-type highly abundant in skin. Blue light irradiation at low doses (<300 mJ cm-2) triggers synthesis of hydrogen peroxide (H2O2) in T cells revealed by the genetically encoded reporter HyPerRed. In turn, H2O2 activates a Src kinase/phospholipase C-γ1 (PLC-γ1) signaling pathway and Ca2+ mobilization. Pharmacologic inhibition or genetic disruption of Lck kinase, PLC-γ1 or the T cell receptor complex inhibits light-evoked Ca2+ transients. Notably, both light and H2O2 enhance T-cell motility in a Lck-dependent manner. Thus, T lymphocytes possess intrinsic photosensitivity and this property may enhance their motility in skin.


Subject(s)
Cell Movement/radiation effects , Skin/radiation effects , T-Lymphocytes/cytology , T-Lymphocytes/radiation effects , Animals , Calcium/chemistry , Cell Proliferation , Chemotaxis , Humans , Hydrogen Peroxide , Jurkat Cells , Mice , Phospholipase C gamma/metabolism , Phosphorylation , Photons , RNA Interference , Signal Transduction/drug effects , Spleen/cytology , Sunlight , Ultraviolet Rays
14.
Oncotarget ; 7(36): 57932-57942, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27462771

ABSTRACT

Zoledronic acid, a potent nitrogen-containing bisphosphonate (NBP), has been extensively used to limit bone turnover in a various diseases including tumors. Recent clinical studies have demonstrated direct anti-cancer effects of zoledronic acid, in addition to its clinical benefits for skeletal-related events. Here we investigated the effects of 4 clinically available NBPs on human tumor cell proliferation. Our data demonstrate a potent anti-proliferative effect of zoledronic acid against glioblastoma (GBM) cell lines, breast cancer cells and GBM patient-derived lines. Zoledronic acid also effectively inhibited GBM tumor growth in xenograft mouse models. Zoledronic acid strongly stimulated autophagy but not apoptotic signals in all tested cells. Only one intermediate product of cholesterols synthesis pathway, geranylgeranyl diphosphate (GGPP) rescued cells from the cytotoxic effects of zoledronic acid. To further investigate the effect of GGPP, we knocked down RABGGTA, which encodes a subunit of the Rabgeranylgeranyltransferase protein. This knockdown induced an effect similar to zoledronic acid in cancer cell lines. These data are promising and suggested a potential for zoledronic acid as an anti-cancer agent, through its ablation of the function of Rab proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Diphosphonates/pharmacology , Nitrogen/chemistry , Animals , Antineoplastic Agents/chemistry , Autophagy , Bone Density Conservation Agents/pharmacology , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Diphosphonates/chemistry , Drug Screening Assays, Antitumor , Glioblastoma/drug therapy , Humans , Imidazoles/chemistry , MCF-7 Cells , Mice , Neoplasm Transplantation , Zoledronic Acid
15.
Oncotarget ; 7(16): 21556-69, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-26933808

ABSTRACT

Glioblastomas (GBM) are the most aggressive and prevalent form of gliomas with abysmal prognosis and limited treatment options. We analyzed clinically relevant molecular aberrations suggestive of response to therapies in 1035 GBM tumors. Our analysis revealed mutations in 39 genes of 48 tested. IHC revealed expression of PD-L1 in 19% and PD-1 in 46%. MGMT-methylation was seen in 43%, EGFRvIII in 19% and 1p19q co-deletion in 2%. TP53 mutation was associated with concurrent mutations, while IDH1 mutation was associated with MGMT-methylation and TP53 mutation and was mutually exclusive of EGFRvIII mutation. Distinct biomarker profiles were seen in GBM compared with WHO grade III astrocytoma, suggesting different biology and potentially different treatment approaches. Analysis of 17 metachronous paired tumors showed frequent biomarker changes, including MGMT-methylation and EGFR aberrations, indicating the need for a re-biopsy for tumor profiling to direct subsequent therapy. MGMT-methylation, PR and TOPO1 appeared as significant prognostic markers in sub-cohorts of GBM defined by age. The current study represents the largest biomarker study on clinical GBM tumors using multiple technologies to detect gene mutation, amplification, protein expression and promoter methylation. These data will inform planning for future personalized biomarker-based clinical trials and identifying effective treatments based on tumor biomarkers.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , DNA Methylation , Gene Amplification , Glioblastoma/genetics , Mutation , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Middle Aged , Prognosis , Promoter Regions, Genetic/genetics , Survival Analysis , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Young Adult
16.
J Transl Med ; 13: 269, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26283544

ABSTRACT

BACKGROUND: The STAT3 transcription factor is a major intracellular signaling protein and is frequently dysregulated in the most common and lethal brain malignancy in adults, glioblastoma multiforme (GBM). Activation of STAT3 in GBM correlates with malignancy and poor prognosis. The phosphorylating signal transducer JAK2 activates STAT3 in response to cytokines and growth factors. Currently there are no JAK-STAT pathway inhibitors in clinical trials for GBM, so we sought to examine the anti-GBM activity of SAR317461 (Sanofi-Aventis), a newer generation, highly potent JAK2 inhibitor that exhibits low toxicity and good pharmacokinetics. SAR317461 was initially approved for patient testing in the treatment of primary myelofibrosis (PMF), and has shown activity in preclinical models of melanoma and pulmonary cancer, but has not been tested in GBM. METHODS: We hypothesized that a potent small molecule JAK2 inhibitor could overcome the heterogeneous nature of GBM, and suppress a range of patient derived GBM tumorsphere lines and immortalized GBM cell lines. We treated with SAR317461 to determine IC50 values, and using Western blot analysis we asked whether the response was linked to STAT3 expression. Western blot analysis, FACS, and cell viability studies were used to identify the mechanism of SAR317461 induced cell death. RESULTS: We report for the first time that the JAK2 inhibitor SAR317461 clearly inhibited STAT3 phosphorylation and had substantial activity against cells (IC50 1-10 µM) from 6 of 7 different patient GSC derived GBM tumorsphere lines and three immortalized GBM lines. One patient GSC derived line did not constitutively express STAT3 and was more resistant to SAR317461 (IC50 ≈25 µM). In terms of mechanism we found cleaved PARP and clear apoptosis following SAR317461. SAR317461 also induced autophagy and the addition of an autophagy inhibitor markedly enhanced cell killing by SAR317461. CONCLUSIONS: We conclude that SAR317461 potently inhibits STAT3 phosphorylation and that it has significant activity against those GBM cells which express activated STAT3. Further studies are warranted in terms of the potential of SAR317461 as single and combined therapy for selectively treating human patients afflicted with GBMs expressing activation of the JAK2-STAT3 signaling axis.


Subject(s)
Brain Neoplasms/metabolism , Enzyme Inhibitors/chemistry , Glioblastoma/metabolism , Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/metabolism , Pyrimidines/chemistry , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/chemistry , Adolescent , Adult , Aged , Autophagy , Brain Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Separation , Cell Survival , Down-Regulation , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Humans , Inhibitory Concentration 50 , Male , Middle Aged , Phosphorylation , Proportional Hazards Models , Young Adult
17.
Expert Rev Anticancer Ther ; 15(8): 955-62, 2015.
Article in English | MEDLINE | ID: mdl-26118895

ABSTRACT

Gliomas are the most common primary malignant brain tumor. Over the last decade, significant advances have been made in the molecular characterization of this tumor group, identifying predictive biomarkers or molecular actionable targets, and paving the way to molecular-based targeted therapies. This personalized therapeutic approach is effective and illustrated in the present review. Among many molecular abnormalities, BRAF mutation and mTOR activation in pilocytic astrocytomas and subependymal giant cell astrocytomas are actionable targets sensitive to vemurafenib and everolimus, respectively. Chromosome arms 1p/19q co-deletion and IDH mutational status are pivotal in driving delivery of early procarbazine, lomustine and vincristine chemotherapy in anaplastic oligodendroglial tumors. Although consensus to assess MGMT promoter methylation is not reached yet, it may be useful in predicting resistance to temozolomide in elderly patients.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Molecular Targeted Therapy , Aged , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Drug Resistance, Neoplasm , Glioma/genetics , Glioma/pathology , Humans , Precision Medicine/methods
18.
Oncotarget ; 6(9): 7293-304, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25762646

ABSTRACT

Invasive growth is a major determinant of the high lethality of malignant gliomas. Plexin-B2, an axon guidance receptor important for mediating neural progenitor cell migration during development, is upregulated in gliomas, but its function therein remains poorly understood. Combining bioinformatic analyses, immunoblotting and immunohistochemistry of patient samples, we demonstrate that Plexin-B2 is consistently upregulated in all types of human gliomas and that its expression levels correlate with glioma grade and poor survival. Activation of Plexin-B2 by Sema4C ligand in glioblastoma cells induced actin-based cytoskeletal dynamics and invasive migration in vitro. This proinvasive effect was associated with activation of the cell motility mediators RhoA and Rac1. Furthermore, costimulation of Plexin-B2 and the receptor tyrosine kinase Met led to synergistic Met phosphorylation. In intracranial glioblastoma transplants, Plexin-B2 knockdown hindered invasive growth and perivascular spreading, and resulted in decreased tumor vascularity. Our results demonstrate that Plexin-B2 promotes glioma invasion and vascularization, and they identify Plexin-B2 as a potential novel prognostic marker for glioma malignancy. Targeting the Plexin-B2 pathway may represent a novel therapeutic approach to curtail invasive growth of glioblastoma.


Subject(s)
Brain Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Nerve Tissue Proteins/metabolism , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement , Computational Biology , GTP Phosphohydrolases/metabolism , Gene Expression Profiling , Glioblastoma , Humans , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , Phosphorylation , Semaphorins/metabolism , Up-Regulation , rho-Associated Kinases/metabolism
19.
Mol Biosyst ; 11(4): 1012-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25648506

ABSTRACT

Glioblastoma is a highly-aggressive and rapidly-lethal tumor characterized by resistance to therapy. Although data on multiple genes, proteins, and pathways are available, the key challenge is deciphering this information and identifying central molecular targets. Therapeutically targeting individual molecules is often unsuccessful due to the presence of compensatory and redundant pathways, and crosstalk. A systems biology approach that involves a hierarchical gene group networks analysis can delineate the coherent functions of different disease mediators. Here, we report an integrative networks-based analysis to identify a system of coherent gene modules in primary and secondary glioblastoma. Our study revealed a hierarchical transcriptional control of genes in these modules. We elucidated those modules responsible for conversion of the glioma-associated microglia/macrophages into glioma-supportive, immunosuppressive cells. Further, we identified clusters comprising mediators of angiogenesis, proliferation, and cell death for both primary and secondary glioblastomas. Data obtained for these clusters point to a possible role of transcription regulators that function as the gene modules mediators in glioblastoma pathogenesis. We elucidated a set of possible transcription regulators that can be targeted to affect the selected gene clusters at specific levels for glioblastoma. Our innovative approach to construct informative disease models may hold the key to successful management of complex diseases including glioblastoma and other cancers.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioblastoma/genetics , Multigene Family/genetics , Brain/metabolism , Brain Neoplasms/metabolism , Cell Death/genetics , Cell Proliferation/genetics , Gene Expression Profiling , Glioblastoma/metabolism , Humans , Neovascularization, Pathologic/genetics , Oligonucleotide Array Sequence Analysis
20.
Oncotarget ; 6(2): 1157-70, 2015 Jan 20.
Article in English | MEDLINE | ID: mdl-25528767

ABSTRACT

Metabolic reprogramming is a key feature of tumorigenesis that is controlled by oncogenes. Enhanced utilization of glucose and glutamine are the best-established hallmarks of tumor metabolism. The oncogene c-Myc is one of the major players responsible for this metabolic alteration. However, the molecular mechanisms involved in Myc-induced metabolic reprogramming are not well defined. Here we identify p32, a mitochondrial protein known to play a role in the expression of mitochondrial respiratory chain complexes, as a critical player in Myc-induced glutamine addiction. We show that p32 is a direct transcriptional target of Myc and that high level of Myc in malignant brain cancers correlates with high expression of p32. Attenuation of p32 expression reduced growth rate of glioma cells expressing Myc and impaired tumor formation in vivo. Loss of p32 in glutamine addicted glioma cells induced resistance to glutamine deprivation and imparted sensitivity to glucose withdrawal. Finally, we provide evidence that p32 expression contributes to Myc-induced glutamine addiction of cancer cells. Our findings suggest that Myc promotes the expression of p32, which is required to maintain sufficient respiratory capacity to sustain glutamine metabolism in Myc transformed cells.


Subject(s)
Brain Neoplasms/genetics , Carrier Proteins/genetics , Glioma/genetics , Glutamine/metabolism , Mitochondrial Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Glioma/pathology , Humans , Immunoblotting , Immunohistochemistry , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mitochondrial Proteins/metabolism , Models, Genetic , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...