• 제목/요약/키워드: Glioblastoma tumor

검색결과 126건 처리시간 0.022초

각종 뇌 종양의 Thallium-201 뇌 SPECT에서 Thallium-201의 동태 (Thallium-201 Uptake and Washout in T1-201 Brain SPECT of Various Brain Tumors)

  • 임상무;홍성운;이창훈;이승훈;김종현
    • 대한핵의학회지
    • /
    • 제26권2호
    • /
    • pp.360-364
    • /
    • 1992
  • Treatment for the brain tumors consist of surgery, chemotherapy, and a variety of methods of irradiation. Therapy is aimed to destroy the tumor, but necrosis and edema occur concurrently. Conventional structural imaging techniques such as CT or MRI are unable to reliably distinguish persistent and recurrent tumor from necrosis or edema. T1-201 has been shown to be useful in the evaluation of the myocardial viability by comparing the early uptake and redistribution image. The aim of this study is to evaluate the clinical usefulness of the early uptake and delayed washout images of the T1-201 brain SPECT in the brain tumors. In the pathologically diagnosed various brain tumor patients, brain SPECT was done with rotating gamma camera 15 minutes and 3 hours after T1-201 injection, and the T1-201 uptake in the tumor was compared with the skull and scalp activity. In the glioblastoma multiforme, meningioma and metastatic tumor, the T1-201 uptake was higher than low grade glioma in both 15 minute and 3 hour images (p<0.02). In the low grade glioma,3 hour T1-201 uptake was significantly lower than 15 minute uptake (p<0.05) but in the glioblastoma, meningioma and metastatic tumor there was no significant difference. There was no significant difference in the T1-201 uptake among the glioblastoma, meningioma and metastatic tumors. In one matastatic tumor, T1-201 uptake was decreased after radiation therapy. T1-201 brain SPECT could distinguish the benign and malignancy, and seems to be useful in the follow-up after treatment. But one of the early or delayed SPECT seems not to be necessary for these purposes.

  • PDF

Ketone ester supplementation of Atkins-type diet prolongs survival in an orthotopic xenograft model of glioblastoma

  • Hassan Azari;Angela Poff;Dominic D'Agostino;Brent Reynolds
    • Anatomy and Cell Biology
    • /
    • 제57권1호
    • /
    • pp.97-104
    • /
    • 2024
  • Heavy reliance on glucose metabolism and a reduced capacity to use ketone bodies makes glioblastoma (GBM) a promising candidate for ketone-based therapies. Ketogenic diet (KD) is well-known for its promising effects in controlling tumor growth in GBM. Moreover, synthetic ketone ester (KE) has demonstrated to increase blood ketone levels and enhance animal survival in a metastatic VM-M3 murine tumor model. Here, we compared the efficacy of a KE-supplemented Atkins-type diet (ATD-KE) to a classic KD in controlling tumor progression and enhancing survival in a clinically relevant orthotopic patient-derived xenograft GBM model. Our findings demonstrate that ATD-KE preserves body weight (percent change from the baseline; 112±2.99 vs. 116.9±2.52 and 104.8±3.67), decreases blood glucose (80.55±0.86 vs. 118.6±9.51 and 52.35±3.89 mg/dl), and increases ketone bodies in blood (1.15±0.03 mM vs. 0.55±0.04 and 2.66±0.21 mM) and brain tumor tissue (3.35±0.30 mM vs. 2.04±0.3 and 4.25±0.25 mM) comparable to the KD (results presented for ATD-KE vs. standard diet [STD] and KD, respectively). Importantly, the ATD-KE treatment significantly enhanced survival compared to the STD and was indistinguishable from the KD (47 days in STD vs. 56 days in KD and ATD-KE), suggesting that a nutritionally balanced low carbohydrate ATD combined with KE may be as effective as the KD alone in reducing brain tumor progression. Overall, these data support the rationale for clinical testing of KE-supplemented low-carb diet as an adjunct treatment for brain tumor patients.

The Value of Tumor Treating Fields in Glioblastoma

  • Zhang, Chaochao;Du, Jianyang;Xu, Weidong;Huang, Haiyan;Gao, Li
    • Journal of Korean Neurosurgical Society
    • /
    • 제63권6호
    • /
    • pp.681-688
    • /
    • 2020
  • Glioblastoma (GBM) is one of the most common tumors of the central nervous system, which is the most lethal brain cancer. GBM treatment is based primarily on surgical resection, combined with radiotherapy and chemotherapy. Despite the positive treatment, progression free survival and overall survival were not significantly prolonged because GBM almost always recurs. We are always looking forward to some new and effective treatments. In recent years, a novel treatment method called tumor treating fields (TTFields) for cancer treatment has been proposed. TTFields devices were approved by the Food and Drug Administration (FDA) for adjuvant treatment of recurrent and newly diagnosed GBMs in 2011 and 2015, respectively. This became the first breakthrough treatment for GBM in the past 10 years after the FDA approved bevacizumab for patients with relapsed GBM in 2009. This paper summarized the research results of TTFields in recent years and elaborated the mechanism of action of TTFields on GBM, including cell and animal experimental research, clinical application and social benefits.

Midline Glioblastoma Multiforme With Bilateral Symmetric Cysts

  • Lee, Hai-Ong;Koh, Eun-Jeong;Oh, Young-Min;Choi, Ha-Young
    • Journal of Korean Neurosurgical Society
    • /
    • 제43권2호
    • /
    • pp.105-108
    • /
    • 2008
  • Cystic glioblastoma multiforme (GBM) is a rare disease. Its exact prevalence has not yet been reported. Also, the mechanism of cyst formation remains to be elucidated. We report a case of GBM with a large peripheral cyst. A 43-year-old woman visited our clinic with a 3-month history of severe headache, memory impairment and general weakness. T1-weighted gadolinium-enhanced magnetic resonance (MR) image revealed a midline enhanced solid mass and bilateral symmetric banana-shaped peripheral cysts. A centrally enhanced mass was measured $2{\times}4$ cm in size and both mass and cysts as $7{\times}7$ cm. Both the frontal lobe and the frontal horn were severely compressed inferiorly and posteriorly. We resected a midline solid tumor and cysts via the bilateral interhemispheric transcortical approach. Histopathologic examination revealed GBM. The patient was subsequently treated with fractionated conventional brain radiation therapy, followed by temozolomide chemotherapy. Eighteen months later, there was no tumor recurrence and no neurological deficits were noted. Our patient showed no tumor recurrence and a long survival at a long follow-up.

Peptide Micelles for Anti-cancer Drug Delivery in an Intracranial Glioblastoma Animal Model

  • Yi, Na;Lee, Minhyung
    • Bulletin of the Korean Chemical Society
    • /
    • 제35권10호
    • /
    • pp.3030-3034
    • /
    • 2014
  • Bis-chloroethylnitrosourea (BCNU) is currently used as an anti-cancer drug for glioblastoma therapy. In this study, BCNU was loaded into the hydrophobic cores of R3V6 amphiphilic peptide micelles for efficient delivery into brain tumors. The scanning electron microscope (SEM) study showed that the BCNU-loaded R3V6 peptide micelles (R3V6-BCNU) formed spherical micelles. MTT assay showed that R3V6-BCNU more efficiently induced cell death in C6 glioblastoma cells than did BCNU. In the Annexin V assay, R3V6-BCNU more efficiently induced apoptosis than did BCNU alone. Furthermore, the results showed that R3V6 was not toxic to cells. The positive charges of the R3V6 peptide micelles may facilitate the interaction between R3V6-BCNU and the cellular membrane, resulting in an increase in cellular uptake of BCNU. In vivo evaluation with an intracranial glioblastoma rat model showed that R3V6-BCNU more effectively reduced tumor size than BCNU alone. The results suggest that R3V6 peptide micelles may be an efficient carrier of BCNU for glioblastoma therapy.

한양방결합치료로 장기간 생존하고 있는 교모세포종 증례 보고 (Case Report on Long Term Survival of Glioblastoma Patient Treated with Integrative Medicine)

  • 양주노;윤영주;홍진우;한창우;권정남;이인;박성하;김소연;최준용;이혜윤
    • 대한한의학회지
    • /
    • 제39권1호
    • /
    • pp.75-85
    • /
    • 2018
  • 교모세포종 진단을 받고 항암치료와 수술을 시행한 30대 남자에게 서양의학, 한의학적인 치료를 결합하여 시행한 결과, 현재 비록 치료 후유증인 편마비가 남아 있으나 이미 14년간 재발없이 장기간 생존함으로써 한방치료의 가능성을 보여 주었기에, 이에 증례를 보고한다.

Parathyroid Hormone-Related Protein Promotes the Proliferation of Patient-Derived Glioblastoma Stem Cells via Activating cAMP/PKA Signaling Pathway

  • Zhenyu Guo;Tingqin Huang;Yingfei Liu;Chongxiao Liu
    • International Journal of Stem Cells
    • /
    • 제16권3호
    • /
    • pp.315-325
    • /
    • 2023
  • Background and Objectives: Glioblastoma (GBM) is an aggressive primary brain tumor characterized by its heterogeneity and high recurrence and lethality rates. Glioblastoma stem cells (GSCs) play a crucial role in therapy resistance and tumor recurrence. Therefore, targeting GSCs is a key objective in developing effective treatments for GBM. The role of Parathyroid hormone-related peptide (PTHrP) in GBM and its impact on GSCs remains unclear. This study aimed to investigate the effect of PTHrP on GSCs and its potential as a therapeutic target for GBM. Methods and Results: Using the Cancer Genome Atlas (TCGA) database, we found higher expression of PTHrP in GBM, which correlated inversely with survival. GSCs were established from three human GBM samples obtained after surgical resection. Exposure to recombinant human PTHrP protein (rPTHrP) at different concentrations significantly enhanced GSCs viability. Knockdown of PTHrP using target-specific siRNA (siPTHrP) inhibited tumorsphere formation and reduced the number of BrdU-positive cells. In an orthotopic xenograft mouse model, suppression of PTHrP expression led to significant inhibition of tumor growth. The addition of rPTHrP in the growth medium counteracted the antiproliferative effect of siPTHrP. Further investigation revealed that PTHrP increased cAMP concentration and activated the PKA signaling pathway. Treatment with forskolin, an adenylyl cyclase activator, nullified the antiproliferative effect of siPTHrP. Conclusions: Our findings demonstrate that PTHrP promotes the proliferation of patient-derived GSCs by activating the cAMP/PKA signaling pathway. These results uncover a novel role for PTHrP and suggest its potential as a therapeutic target for GBM treatment.

Kaposi's Sarcoma-Associated Herpesvirus Infection Modulates the Proliferation of Glioma Stem-Like Cells

  • Jeon, Hyungtaek;Kang, Yun Hee;Yoo, Seung-Min;Park, Myung-Jin;Park, Jong Bae;Lee, Seung-Hoon;Lee, Myung-Shin
    • Journal of Microbiology and Biotechnology
    • /
    • 제28권1호
    • /
    • pp.165-174
    • /
    • 2018
  • Glioblastoma multiforme is the most lethal malignant brain tumor. Despite many intensive studies, the prognosis of glioblastoma multiforme is currently very poor, with a median overall survival duration of 14 months and 2-year survival rates of less than 10%. Although viral infections have been emphasized as potential cofactors, their influences on pathways that support glioblastoma progression are not known. Some previous studies indicated that human Kaposi's sarcoma-associated herpesvirus (KSHV) was detected in healthy brains, and its microRNA was also detected in glioblastoma patients' plasma. However, a direct link between KSHV infection and glioblastoma is currently not known. In this study, we infected glioblastoma cells and glioma stem-like cells (GSCs) with KSHV to establish an in vitro cell model for KSHV-infected glioblastoma cells and glioma stem-like cells in order to identify virologic outcomes that overlap with markers of aggressive disease. Latently KSHV-infected glioblastoma cells and GSCs were successfully established. Additionally, using these cell models, we found that KSHV infection modulates the proliferation of glioma stem-like cells.

AntagomiR-27a Targets FOXO3a in Glioblastoma and Suppresses U87 Cell Growth in Vitro and in Vivo

  • Ge, Yun-Fei;Sun, Jun;Jin, Chun-Jie;Cao, Bo-Qiang;Jiang, Zhi-Feng;Shao, Jun-Fei
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권2호
    • /
    • pp.963-968
    • /
    • 2013
  • Objective: To study the effect of the antagomiR-27a inhibitor on glioblastoma cells. Methods: The miR-27a expression level in specimens of human glioblastoma and normal human brain tissues excised during decompression for traumatic brain injury was assessed using qRT-PCR; The predicted target gene of miR-27a was screened out through bioinformatics databases, and the predicted gene was verified using genetic report assays; the effect of antagomiR-27a on the invasion and proliferation of glioma cells was analyzed using MTT assays and 5-ethynyl-2'-deoxyuridine (EdU) labeling. A xenograft glioblastoma model in BALB-c nude mice was established to detect the effect of antagomiR-27a on tumour growth. Results: qRT-PCR results showed that miR-27a significantly increased in specimens from glioblastoma comparing with normal human brain tissues. Th miR-27a inhibitor significantly suppressed invasion and proliferation of glioblastoma cells. FOXO3a was verified as a new target of miR-27a by Western blotting and reporter analyzes. Tumor growth in vivo was suppressed by administration of the miR-27a inhibitor. Conclusion: MiR-27a may be up-regulated in human glioblastoma, and antagomiR-27a could inhibit the proliferation and invasion ability of glioblastoma cells.

Propranolol Inhibits the Proliferation of Human Glioblastoma Cell Lines through Notch1 and Hes1 Signaling System

  • Kim, Hyun Sik;Park, Young Han;Lee, Heui Seung;Kwon, Mi Jung;Song, Joon Ho;Chang, In Bok
    • Journal of Korean Neurosurgical Society
    • /
    • 제64권5호
    • /
    • pp.716-725
    • /
    • 2021
  • Objective : The anti-tumor effect of the beta-adrenergic receptor antagonist propranolol in breast cancer is well known; however, its activity in glioblastoma is not well-evaluated. The Notch-Hes pathway is known to regulate cell differentiation, proliferation, and apoptosis. We investigated the effect of propranolol to human glioblastoma cell lines, and the role of Notch and Hes signaling in this process. Methods : We performed immunohistochemical staining on 31 surgically resected primary human glioblastoma tissues. We also used glioblastoma cell lines of U87-MG, LN229, and neuroblastoma cell line of SH-SY5Y in this study. The effect of propranolol and isoproterenol on cell proliferation was evaluated using the MTT assay (absorbance 570 nm). The impact of propranolol on gene expression (Notch and Hes) was evaluated using real-time polymerase chain reaction (RT-PCR, whereas protein levels of Notch1 and Hes1 were measured using Western blotting (WB), simultaneously. Small interfering RNA (siRNA) was used to suppress the Notch gene to investigate its role in the proliferation of glioblastoma. Results : Propranolol and isoproterenol caused a dose-dependent decrease in cell proliferation (MTT assay). RT-PCR showed an increase in Notch1 and Hes1 expression by propranolol, whereas WB demonstrated increase in Notch1 protein, but a decrease in Hes1 by propranolol. The proliferation of U87-MG and LN229 was not significantly suppressed after transfection with Notch siRNA. Conclusion : These results demonstrated that propranolol suppressed the proliferation of glioblastoma cell lines and neuroblastoma cell line, and Hes1 was more closely involved than Notch1 was in glioblastoma proliferation.