Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
World Neurosurg ; 148: e406-e414, 2021 04.
Article in English | MEDLINE | ID: mdl-33444828

ABSTRACT

OBJECTIVE: The relationship between quantitative posturography results and growth of vestibular schwannomas (VSs) during conservative management has not been studied. We aimed to clarify the relationship between the presence of disequilibrium based on posturographic measurement and VS growth. METHODS: This retrospective, single-center study included 53 patients with VSs (Koos stage I or II) managed conservatively after initial diagnosis. Radiographic progression was considered present if 20% volumetric growth was observed over the imaging interval. Posturography was performed at initial diagnosis, and sway velocity (SV) and sway area were calculated. Tumor growth-free survival was estimated using the Kaplan-Meier method. RESULTS: Mean follow-up period was 2.87 ± 2.58 years, up to tumor growth detection or last follow-up magnetic resonance imaging. Tumor growth incidence was 40.8% and 61.2% at 2 and 5 years, respectively. Cerebellopontine angle extension and SV with eyes open were related to tumor growth. Tumor growth-free survival of patients with cerebellopontine angle extension and patients with intracanalicular tumor at 2 years was 37.3% and 76.4%, respectively. Tumor growth-free survival of patients with SV >2.06 cm/second and patients with SV ≤2.06 cm/second at 2 years was 30.8% and 68.9%, respectively. The Cox hazard model demonstrated a significant risk for future tumor growth with SV >2.06 cm/second (relative risk, 2.475; 95% confidence interval, 1.11-5.37, P = 0.027). CONCLUSIONS: We demonstrated a positive correlation between SV with eyes open and future tumor growth. Posturographic data are objective and quantitative; thus, SV may be a potential predictor of future growth of VSs.


Subject(s)
Neuroma, Acoustic/pathology , Postural Balance/physiology , Sensation Disorders/etiology , Aged , Conservative Treatment , Female , Follow-Up Studies , Hearing Loss, Unilateral/etiology , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Neuroma, Acoustic/complications , Neuroma, Acoustic/diagnostic imaging , Neuroma, Acoustic/therapy , Prognosis , Progression-Free Survival , Radiosurgery , Retrospective Studies , Tumor Burden
2.
NMC Case Rep J ; 8(1): 129-135, 2021.
Article in English | MEDLINE | ID: mdl-35079454

ABSTRACT

von Hippel-Lindau (VHL) disease is characterized by neoplastic and cystic lesions, such as central nervous system (CNS) hemangioblastoma and clear cell renal cell carcinoma (RCC), arising in multiple organs. Here, we report a case of an RCC that metastasized to a spinal hemangioblastoma in a patient diagnosed with VHL disease. This is a unique case study because visceral neoplasms rarely metastasize to the CNS. The patient had undergone posterior fossa surgery for the removal of hemangioblastomas in the right cerebellar hemisphere as a child. He was diagnosed with RCC at the age of 20 years, and he underwent partial nephrectomy at the age of 35 years. The patient underwent surgical removal of a spinal tumor from Th8, which was also diagnosed as a hemangioblastoma at the age of 40. However, the residual spinal tumor rapidly regrew within 1.5 years. A second surgery was performed due to progressive leg motor weakness. The resected tumor from the second surgery had two distinct components between the tumor center and the margin. Immunohistochemistry of CD10, PAX 8, and inhibin A demonstrated the predominant region of the tumor was RCC. Pathological findings confirmed tumor-to-tumor metastasis of the RCC migrating into residual spinal hemangioblastoma. It can be challenging to distinguish hemangioblastoma from RCC in neuroimaging. We suggest that tumor-to-tumor metastasis should be considered as a differential diagnosis if benign tumors grow rapidly, even if the pathological diagnosis does not initially confirm malignancy. The biological mechanisms of RCC migrating into residual hemangioblastoma are discussed.

4.
J Biol Chem ; 294(51): 19740-19751, 2019 12 20.
Article in English | MEDLINE | ID: mdl-31712311

ABSTRACT

In cancer, aberrant growth factor receptor signaling reprograms cellular metabolism and global gene transcription to drive aggressive growth, but the underlying mechanisms are not well-understood. Here we show that in the highly lethal brain tumor glioblastoma (GBM), mTOR complex 2 (mTORC2), a critical core component of the growth factor signaling system, couples acetyl-CoA production with nuclear translocation of histone-modifying enzymes including pyruvate dehydrogenase and class IIa histone deacetylases to globally alter histone acetylation. Integrated analyses in orthotopic mouse models and in clinical GBM samples reveal that mTORC2 controls iron metabolisms via histone H3 acetylation of the iron-related gene promoter, promoting tumor cell survival. These results nominate mTORC2 as a critical epigenetic regulator of iron metabolism in cancer.


Subject(s)
Brain Neoplasms/metabolism , Epigenesis, Genetic , Glioblastoma/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Iron/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Active Transport, Cell Nucleus , Animals , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Histones/chemistry , Humans , Immediate-Early Proteins/metabolism , Metabolome , Mice , Neoplasm Transplantation , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Signal Transduction
5.
World Neurosurg ; 129: e733-e740, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31285176

ABSTRACT

BACKGROUND: The surgical resection of large supracerebellar hemangioblastomas (SHBs) is exceptionally challenging due to their vascularity and deep anatomic location and is associated with a high risk of postoperative complications and mortality. Access to the posterior incisural space can be achieved by either an infratentorial supracerebellar approach or occipital transtentorial approach (OTA). However, the optimal surgical strategy has not yet been established. Here, we report 2 cases of large SHBs that were successfully and safely resected via a unilateral OTA with multimodal assistance. CASE DESCRIPTION: Two patients presented to our hospital with ataxia due to large, solid SHBs. After preoperative embolization, gross total resection of the SHBs was achieved via an OTA. Furthermore, endoscopic assistance was used to resect the remnant portion of the tumor in the second patient. Both patients experienced transient ataxia but were discharged from the hospital without serious complications. CONCLUSIONS: The combination of an OTA with preoperative embolization and endoscopic assistance may reduce the intraoperative risk and contribute to improved outcome in patients with such clinically challenging tumors.


Subject(s)
Brain Neoplasms/surgery , Hemangioblastoma/surgery , Neurosurgical Procedures/methods , Aged , Brain Neoplasms/diagnostic imaging , Embolization, Therapeutic , Female , Hemangioblastoma/diagnostic imaging , Humans , Magnetic Resonance Imaging , Neuroendoscopy , Treatment Outcome
6.
World Neurosurg ; 130: 71-76, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31279920

ABSTRACT

BACKGROUND: Cerebellar cyst formation after surgery is uncommon, and few cases of this condition have been previously reported. These cases had an intraparenchymal cyst in the cerebellar hemisphere that required surgical fenestration of the cyst. We herein present a rare case of a postoperative cerebellar cyst with pseudomeningocele and magnetic resonance images indicating a fistula between the cyst and pseudomeningocele. CASE DESCRIPTION: A patient presented with an intraparenchymal cyst and surrounding edema in the cerebellar hemisphere that developed after a C1 laminectomy and a small suboccipital craniectomy for the removal of an accessory nerve neurinoma at the craniovertebral junction. Fast imaging employing steady-state acquisition images identified the fistula connecting the cyst and extradural cerebrospinal fluid retention. Conservative management with administration of dexamethasone induced spontaneous regression of the cyst, and no recurrence had occurred by the 1-year follow-up. CONCLUSIONS: Watertight dural closure is important for the prevention of this rare complication after posterior fossa surgery. However, an arachnoid tear on the cerebellar fissure and adjacent dural defect are necessary antecedents for this rare condition. High-resolution fast imaging employing steady-state acquisition images could provide additional information for the etiology of postoperative cerebellar cyst.


Subject(s)
Central Nervous System Cysts/diagnostic imaging , Cerebellum/diagnostic imaging , Meningocele/diagnostic imaging , Postoperative Complications/diagnostic imaging , Spinal Neoplasms/diagnostic imaging , Spinal Neoplasms/surgery , Aged , Central Nervous System Cysts/etiology , Cervical Vertebrae/diagnostic imaging , Humans , Male , Meningocele/etiology , Postoperative Complications/etiology , Skull Base/diagnostic imaging
7.
Cell Metab ; 30(3): 525-538.e8, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31303424

ABSTRACT

Advances in DNA sequencing technologies have reshaped our understanding of the molecular basis of cancer, providing a precise genomic view of tumors. Complementary biochemical and biophysical perspectives of cancer point toward profound shifts in nutrient uptake and utilization that propel tumor growth and major changes in the structure of the plasma membrane of tumor cells. The molecular mechanisms that bridge these fundamental aspects of tumor biology remain poorly understood. Here, we show that the lysophosphatidylcholine acyltransferase LPCAT1 functionally links specific genetic alterations in cancer with aberrant metabolism and plasma membrane remodeling to drive tumor growth. Growth factor receptor-driven cancers are found to depend on LPCAT1 to shape plasma membrane composition through enhanced saturated phosphatidylcholine content that is, in turn, required for the transduction of oncogenic signals. These results point to a genotype-informed strategy that prioritizes lipid remodeling pathways as therapeutic targets for diverse cancers.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Gene Amplification , Neoplasms/genetics , Neoplasms/metabolism , Oncogenes/genetics , Phospholipids/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , A549 Cells , Animals , Cell Survival/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Genotype , Heterografts , Humans , Mice , Mice, Nude , PC-3 Cells , Signal Transduction/genetics , Transfection
8.
World Neurosurg ; 127: e873-e880, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30954745

ABSTRACT

BACKGROUND: Corpus callosum (CC) infarction has been reported to be rare because of the rich blood supply in the CC. The pathophysiology of CC infarction associated with acute hydrocephalus is unknown. The aim of the present study was to clarify the characteristics and mechanism of CC infarction associated with acute noncommunicating hydrocephalus (ANCH). METHODS: We reviewed clinical the data from all patients who had undergone surgical intervention for ANCH at Chiba University Hospital from January 2008 to March 2018. Patients with vascular lesions, a history of hydrocephalus, and lacking magnetic resonance imaging studies were excluded. The clinical, surgical, and radiological parameters were obtained retrospectively for pathophysiological analysis. RESULTS: A total of 23 patients with ANCH who had undergone surgical intervention and had met the inclusion criteria were included in the present study. Of the 23 patients, 6 (23%) had developed CC infarction. All CC infarctions were located in the splenium. Although no clinical or surgical features were associated with splenial infarction, the radiological parameters of lateral ventricle enlargement and a narrower callosal angle at the posterior commissure and the foramen of Monro were significantly associated with splenial infarction. CONCLUSION: The present study has presented evidence that increased intraventricular pressure by ANCH applied transversely in the splenium will directly induce compression of the superior branch of the posterior callosal artery and pericallosal pial plexus, resulting in splenium-specific infarction in patients with ANCH.


Subject(s)
Corpus Callosum/pathology , Corpus Callosum/surgery , Hydrocephalus/pathology , Hydrocephalus/surgery , Acute Disease , Adolescent , Adult , Aged , Arteries/pathology , Arteries/surgery , Cerebral Infarction/pathology , Cerebral Infarction/surgery , Child , Child, Preschool , Corpus Callosum/blood supply , Female , Humans , Infarction/pathology , Infarction/surgery , Magnetic Resonance Imaging/methods , Male , Middle Aged , Radiology/methods , Young Adult
9.
Oncotarget ; 10(2): 175-183, 2019 Jan 04.
Article in English | MEDLINE | ID: mdl-30719212

ABSTRACT

Glioblastoma (GBM) is the most common malignant brain tumor, and infiltrates into the surrounding normal brain tissue. Induction of a tumor-specific immune response is one of the best methods to obtain tumor-specific cytotoxicity. Photodynamic therapy (PDT) is known to effectively induce an antitumor immune response. We have developed a clinically translatable nanoparticle, liposomally formulated phospholipid-conjugated indocyanine green (LP-iDOPE), applicable for PDT. This nanoparticle accumulates in tumor tissues by the enhanced permeability and retention effect, and releases heat and singlet oxygen to injure cancer cells when activated by near infrared (NIR) light. We assessed the effectiveness of the LP-iDOPE system in brain using the rat 9L glioblastoma model. Treatment with LP-iDOPE and NIR irradiation resulted in significant tumor growth suppression and prolongation of survival. Histopathological examination showed induction of both apoptosis and necrosis and accumulation of CD8+ T-cells and macrophages/microglia accompanied by marked expressions of heat shock protein-70 (HSP70), which was not induced by mild hyperthermia alone at 45° C or an interleukin-2-mediated immune reaction. Notably, the efficacy was lost in immunocompromised nude rats. These results collectively show that the novel nanoparticle LP-iDOPE in combination with NIR irradiation can efficiently induce a tumor-specific immune reaction for malignant gliomas possibly by inducing HSP70 expression which is known to activate antigen-presenting cells through toll-like receptor signaling.

10.
J Neurooncol ; 141(1): 205-211, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30565028

ABSTRACT

INTRODUCTION: Chromosomes 1p/19q co-deletion is a robust molecular marker for the diagnosis of oligodendroglial tumors, and has been included in the 2016 WHO modified classification. Although treatment for oligodendroglioma is controversial, upfront chemotherapy is regarded as one of the treatment option for low-grade tumor. We have treated all the 1p/19q co-deleted oligodendrogliomas, both grades II and III, with upfront chemotherapy without conventional radiotherapy for 20 years. The clinical experience from this trial may be suggestive for understanding of the biological features of oligodendroglioma with 1p/19q co-deletion toward precision medicine. METHODS: This is a long-term retrospective data of the non-selected patients with 1p/19q co-deleted oligodendrogliomas uniformly treated with up-front chemotherapy. Seventy consecutive patients (48 with grade II and 22 with grade III tumors) were included. RESULTS: The median follow-up period was 13 years. The 5-, 10-, and 15-year progression-free survival (PFS) rates were 85.7%, 54.8%, and 31.5%, respectively, and the median PFS was 146 months. In most cases, tumor recurrence was remained local and could be controlled by salvage surgery and/or chemotherapy. The 5-, 10-, and 15-year overall survival (OS) rates were 96.8%, 88.7%, and 80.0%, respectively, and the median OS was not reached. These survival data compared favorably with previous large clinical studies employing radiotherapy. Tumor grades based on World Health Organization classification, extent of surgery, and age affected neither PFS nor OS. Most patients were able to return to their premorbid social life. CONCLUSIONS: The long-term results drawn from 20-years of single institution experience show that the patients with 1p/19q co-deleted oligodendrogliomas can be successfully treated with up-front chemotherapy alone without compromising OS.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/mortality , Chromosome Deletion , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Oligodendroglioma/drug therapy , Oligodendroglioma/mortality , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Female , Humans , Male , Middle Aged , Neoplasm Grading , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Progression-Free Survival , Retrospective Studies , Survival Rate
11.
Genes Dev ; 31(12): 1212-1227, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28724615

ABSTRACT

In glioblastoma (GBM), heterogeneous expression of amplified and mutated epidermal growth factor receptor (EGFR) presents a substantial challenge for the effective use of EGFR-directed therapeutics. Here we demonstrate that heterogeneous expression of the wild-type receptor and its constitutively active mutant form, EGFRvIII, limits sensitivity to these therapies through an interclonal communication mechanism mediated by interleukin-6 (IL-6) cytokine secreted from EGFRvIII-positive tumor cells. IL-6 activates a NF-κB signaling axis in a paracrine and autocrine manner, leading to bromodomain protein 4 (BRD4)-dependent expression of the prosurvival protein survivin (BIRC5) and attenuation of sensitivity to EGFR tyrosine kinase inhibitors (TKIs). NF-κB and survivin are coordinately up-regulated in GBM patient tumors, and functional inhibition of either protein or BRD4 in in vitro and in vivo models restores sensitivity to EGFR TKIs. These results provide a rationale for improving anti-EGFR therapeutic efficacy through pharmacological uncoupling of a convergence point of NF-κB-mediated survival that is leveraged by an interclonal circuitry mechanism established by intratumoral mutational heterogeneity.


Subject(s)
Drug Resistance, Neoplasm/genetics , Glioblastoma/physiopathology , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction/genetics , Animals , Cell Communication , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Interleukin-6/metabolism , Mice , Mice, Nude , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Mol Cell ; 67(1): 128-138.e7, 2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28648777

ABSTRACT

Mutations in cancer reprogram amino acid metabolism to drive tumor growth, but the molecular mechanisms are not well understood. Using an unbiased proteomic screen, we identified mTORC2 as a critical regulator of amino acid metabolism in cancer via phosphorylation of the cystine-glutamate antiporter xCT. mTORC2 phosphorylates serine 26 at the cytosolic N terminus of xCT, inhibiting its activity. Genetic inhibition of mTORC2, or pharmacologic inhibition of the mammalian target of rapamycin (mTOR) kinase, promotes glutamate secretion, cystine uptake, and incorporation into glutathione, linking growth factor receptor signaling with amino acid uptake and utilization. These results identify an unanticipated mechanism regulating amino acid metabolism in cancer, enabling tumor cells to adapt to changing environmental conditions.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain Neoplasms/enzymology , Cysteine/metabolism , Glioblastoma/enzymology , Glutamine/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , A549 Cells , Amino Acid Transport System y+/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/genetics , Glioblastoma/pathology , Glutathione/biosynthesis , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/genetics , Mutation , Phosphorylation , Protein Binding , Proteomics/methods , RNA Interference , Serine , TOR Serine-Threonine Kinases/genetics , Tandem Mass Spectrometry , Time Factors , Transfection , Tumor Microenvironment
13.
Cancer Cell ; 30(5): 683-693, 2016 Nov 14.
Article in English | MEDLINE | ID: mdl-27746144

ABSTRACT

Small-molecule inhibitors targeting growth factor receptors have failed to show efficacy for brain cancers, potentially due to their inability to achieve sufficient drug levels in the CNS. Targeting non-oncogene tumor co-dependencies provides an alternative approach, particularly if drugs with high brain penetration can be identified. Here we demonstrate that the highly lethal brain cancer glioblastoma (GBM) is remarkably dependent on cholesterol for survival, rendering these tumors sensitive to Liver X receptor (LXR) agonist-dependent cell death. We show that LXR-623, a clinically viable, highly brain-penetrant LXRα-partial/LXRß-full agonist selectively kills GBM cells in an LXRß- and cholesterol-dependent fashion, causing tumor regression and prolonged survival in mouse models. Thus, a metabolic co-dependency provides a pharmacological means to kill growth factor-activated cancers in the CNS.


Subject(s)
Brain Neoplasms/drug therapy , Cholesterol/metabolism , Glioblastoma/drug therapy , Indazoles/administration & dosage , Liver X Receptors/metabolism , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Glioblastoma/metabolism , Humans , Indazoles/pharmacology , Mice , Treatment Outcome
14.
Cancer Cell ; 29(4): 563-573, 2016 Apr 11.
Article in English | MEDLINE | ID: mdl-27070703

ABSTRACT

Intratumoral heterogeneity of signaling networks may contribute to targeted cancer therapy resistance, including in the highly lethal brain cancer glioblastoma (GBM). We performed single-cell phosphoproteomics on a patient-derived in vivo GBM model of mTOR kinase inhibitor resistance and coupled it to an analytical approach for detecting changes in signaling coordination. Alterations in the protein signaling coordination were resolved as early as 2.5 days after treatment, anticipating drug resistance long before it was clinically manifest. Combination therapies were identified that resulted in complete and sustained tumor suppression in vivo. This approach may identify actionable alterations in signal coordination that underlie adaptive resistance, which can be suppressed through combination drug therapy, including non-obvious drug combinations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/therapeutic use , Proteomics/methods , Single-Cell Analysis/methods , Adaptation, Physiological , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Butadienes/administration & dosage , Dasatinib/administration & dosage , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/physiology , Gene Expression Profiling , Genes, erbB-1 , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Models, Biological , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/physiology , Mutation , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Nitriles/administration & dosage , Pyrazines/administration & dosage , Selection, Genetic , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/physiology , Xenograft Model Antitumor Assays
15.
Mol Cell ; 60(2): 307-18, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26455392

ABSTRACT

Epidermal growth factor receptor (EGFR) gene amplification and mutations are the most common oncogenic events in glioblastoma (GBM), but the mechanisms by which they promote aggressive tumor growth are not well understood. Here, through integrated epigenome and transcriptome analyses of cell lines, genotyped clinical samples, and TCGA data, we show that EGFR mutations remodel the activated enhancer landscape of GBM, promoting tumorigenesis through a SOX9 and FOXG1-dependent transcriptional regulatory network in vitro and in vivo. The most common EGFR mutation, EGFRvIII, sensitizes GBM cells to the BET-bromodomain inhibitor JQ1 in a SOX9, FOXG1-dependent manner. These results identify the role of transcriptional/epigenetic remodeling in EGFR-dependent pathogenesis and suggest a mechanistic basis for epigenetic therapy.


Subject(s)
Brain Neoplasms/genetics , Epigenesis, Genetic , ErbB Receptors/genetics , Forkhead Transcription Factors/genetics , Glioblastoma/genetics , Nerve Tissue Proteins/genetics , SOX9 Transcription Factor/genetics , Adult , Animals , Azepines/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Child , ErbB Receptors/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Mutation , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , SOX9 Transcription Factor/metabolism , Signal Transduction , Transcriptome , Triazoles/pharmacology
16.
J Neurooncol ; 124(3): 493-500, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26243269

ABSTRACT

Despite accumulating knowledge regarding molecular backgrounds, the optimal management strategy for low-grade gliomas remains controversial. One reason is the marked heterogeneity in the clinical course. To establish an accurate subclassification of low-grade gliomas, we retrospectively evaluated isocitrate dehydrogenase-1 (IDH1) mutation in clinical specimens of diffuse astrocytomas (DA) and oligodendroglial tumors separately. No patients were treated with early radiotherapy, and modified PCV chemotherapy was used for postoperative residual tumors or recurrence in oligodendroglial tumors. Immunohistochemical evaluation of IDH status, p53 status, O(6)-methylguanine methyltransferase expression, and the MIB-1 index were performed. The 1p and 19q status was analyzed with fluorescence in situ hybridization. Ninety-four patients were followed for a median period of 8.5 years. For DAs, p53 was prognostic for progression- free survival (PFS) and IDH1 was significant for overall survival (OS) with multivariate analysis. In contrast, for oligodendroglial tumors, none of the parameters was significant for PFS or OS. Thus, the significance of IDH1 mutation is not clear in oligodendroglial tumors that are homogeneously indolent and chemosensitive. In contrast, DAs are heterogeneous tumors including some potentially malignant tumors that can be predicted by examining the IDH1 mutation status.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Oligodendroglioma/genetics , Radiotherapy/methods , Adult , Age Factors , Aged , Aged, 80 and over , Astrocytoma/radiotherapy , Chromosomes, Human, Pair 19 , Disease-Free Survival , Female , Humans , Karnofsky Performance Status , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Oligodendroglioma/radiotherapy , Prognosis , Retrospective Studies , Treatment Outcome , Tumor Suppressor Protein p53/metabolism , Young Adult
17.
Proc Natl Acad Sci U S A ; 112(30): 9406-11, 2015 Jul 28.
Article in English | MEDLINE | ID: mdl-26170313

ABSTRACT

Cancer cells adapt their signaling in response to nutrient availability. To uncover the mechanisms regulating this process and its functional consequences, we interrogated cell lines, mouse tumor models, and clinical samples of glioblastoma (GBM), the highly lethal brain cancer. We discovered that glucose or acetate is required for epidermal growth factor receptor vIII (EGFRvIII), the most common growth factor receptor mutation in GBM, to activate mechanistic target of rapamycin complex 2 (mTORC2) and promote tumor growth. Glucose or acetate promoted growth factor receptor signaling through acetyl-CoA-dependent acetylation of Rictor, a core component of the mTORC2 signaling complex. Remarkably, in the presence of elevated glucose levels, Rictor acetylation is maintained to form an autoactivation loop of mTORC2 even when the upstream components of the growth factor receptor signaling pathway are no longer active, thus rendering GBMs resistant to EGFR-, PI3K (phosphoinositide 3-kinase)-, or AKT (v-akt murine thymoma viral oncogene homolog)-targeted therapies. These results demonstrate that elevated nutrient levels can drive resistance to targeted cancer treatments and nominate mTORC2 as a central node for integrating growth factor signaling with nutrient availability in GBM.


Subject(s)
Brain Neoplasms/drug therapy , Carrier Proteins/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glucose/chemistry , Acetates/chemistry , Acetyl Coenzyme A/chemistry , Acetylation , Amino Acid Sequence , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Glioblastoma/metabolism , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, SCID , Molecular Sequence Data , Multiprotein Complexes/metabolism , Neoplasm Transplantation , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Sequence Homology, Amino Acid , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
18.
Clin Neurophysiol ; 125(11): 2305-2312, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24731767

ABSTRACT

OBJECTIVE: The P300-based brain-computer interface (BCI) is designed to help patients with motor disabilities to control their environment, and it has been used successfully in patients with amyotrophic lateral sclerosis (ALS). However, some ALS patients were unable to use the visual P300-BCI with the conventional row/column presentation. In this study, we evaluated the effect of a newly developed region-based two-step P300 speller, which has a larger flashing area than the conventional visual array. METHODS: Seven ALS patients and seven age- and sex-matched able-bodied control subjects were required to input hiragana characters using our P300 BCI system. We prepared two types of input procedures, the conventional row/column (RC) speller and the two-step speller, and evaluated their online performance. RESULTS: The mean online accuracy of the ALS patients was 24% for the RC condition and 55% for the two-step condition. The accuracy of the control subjects was 71% and 83% for the RC and two-step condition, respectively. Accuracy in ALS patients was significantly lower than that in the control subjects, and the new visual stimuli significantly increased accuracy of ALS patients. Using the new speller, two ALS patients showed an initial accuracy sufficient for practical use (>70%). The other two ALS patients, who performed better in the first trial using the new speller, continued to experience the BCI system, and their mean accuracy increased to 92%. CONCLUSIONS: The two-step procedure for the visual P300 BCI system provided significantly increased accuracy for ALS patients compared with a conventional RC speller. SIGNIFICANCE: The new region-based two-step P300 speller was effective in ALS patients, and the system may be beneficial to expand their range of activities.


Subject(s)
Amyotrophic Lateral Sclerosis/physiopathology , Brain-Computer Interfaces , Event-Related Potentials, P300/physiology , Aged , Electroencephalography/methods , Female , Humans , Male , Middle Aged
19.
J Neurooncol ; 117(2): 261-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24488444

ABSTRACT

The survival of patients with primary CNS lymphoma (PCNSL) has been improved by high-dose methotrexate (HD-MTX). Since the combination therapy of HD-MTX and whole-brain radiotherapy (WBRT) carries a significant risk for delayed neurotoxicity, it is important to know the therapeutic potential and prognostic factors for HD-MTX without WBRT. We retrospectively reviewed 46 consecutive patients with PCNSL treated with a HD-MTX (3.5 g/m(2)) and deferred WBRT. Patients who achieved complete response or partial response after three courses of HD-MTX were cautiously followed-up without additional treatment. Patients who had either stable disease, progressive disease, or disease relapse were offered salvage therapy. The median progression-free survival period was 10 months and the median overall survival period was 52 months, with a 5-year survival rate of 39 %. Nineteen patients (49 % of the evaluable patients) achieved a complete response at the initial response assessment. Involvement of deep structures of the brain (corpus callosum, basal ganglia and brainstem) was significantly associated with the worse progression-free survivals (p = 0.0058) and overall survivals (p = 0.0177). Gene expression profiling analysis by microarray was compared in eight patients between PCNSLs located in the deep structures of the brain and non-deep-seated tumors. The result showed that up-regulation of signal transduction-related genes and down-regulation of catalytic activity-related genes in the non-deep-seated PCNSL compared with the deep-seated tumors. The present study shows that PCNSL located in non-deep structures of the brain responds better to HD-MTX alone than those involved deep-structures.


Subject(s)
Antineoplastic Agents/administration & dosage , Central Nervous System Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Lymphoma, Non-Hodgkin/drug therapy , Methotrexate/administration & dosage , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Humans , Kaplan-Meier Estimate , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis , Proportional Hazards Models , Retrospective Studies , Transcriptome , Treatment Outcome
20.
Clin Cancer Res ; 19(20): 5722-32, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24030701

ABSTRACT

PURPOSE: mTOR pathway hyperactivation occurs in approximately 90% of glioblastomas, but the allosteric mTOR inhibitor rapamycin has failed in the clinic. Here, we examine the efficacy of the newly discovered ATP-competitive mTOR kinase inhibitors CC214-1 and CC214-2 in glioblastoma, identifying molecular determinants of response and mechanisms of resistance, and develop a pharmacologic strategy to overcome it. EXPERIMENTAL DESIGN: We conducted in vitro and in vivo studies in glioblastoma cell lines and an intracranial model to: determine the potential efficacy of the recently reported mTOR kinase inhibitors CC214-1 (in vitro use) and CC214-2 (in vivo use) at inhibiting rapamycin-resistant signaling and blocking glioblastoma growth and a novel single-cell technology-DNA Encoded Antibody Libraries-was used to identify mechanisms of resistance. RESULTS: Here, we show that CC214-1 and CC214-2 suppress rapamycin-resistant mTORC1 signaling, block mTORC2 signaling, and significantly inhibit the growth of glioblastomas in vitro and in vivo. EGFRvIII expression and PTEN loss enhance sensitivity to CC214 compounds, consistent with enhanced efficacy in strongly mTOR-activated tumors. Importantly, CC214 compounds potently induce autophagy, preventing tumor cell death. Genetic or pharmacologic inhibition of autophagy greatly sensitizes glioblastoma cells and orthotopic xenografts to CC214-1- and CC214-2-induced cell death. CONCLUSIONS: These results identify CC214-1 and CC214-2 as potentially efficacious mTOR kinase inhibitors in glioblastoma, and suggest a strategy for identifying patients most likely to benefit from mTOR inhibition. In addition, this study also shows a central role for autophagy in preventing mTOR-kinase inhibitor-mediated tumor cell death, and suggests a pharmacologic strategy for overcoming it.


Subject(s)
ErbB Receptors/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Glioblastoma/drug therapy , Humans , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Multiprotein Complexes/metabolism , PTEN Phosphohydrolase/metabolism , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...