scholarly journals Rapid Intraoperative Detection of Residual Glioma Cell in Resection Cavity Walls Using a Compact Fluorescence Microscope

Author(s):  
Jiro Akimoto ◽  
shinjiro Fukami ◽  
Megumi Ichikawa ◽  
Kenta nagai ◽  
Michihiro Kohno

Objective: Surgical eradication of malignant glioma cells is theoretically impossible. Therefore, reducing the number of remaining tumor cells around the brain-tumor interface (BTI) is crucial for achieving satisfactory clinical results. The usefulness of fluorescence-guided resection for the treatment of malignant glioma was recently reported, but the detection of infiltrating tumor cells in the BTI using a surgical microscope is not realistic. Therefore, we developed an intraoperative rapid fluorescence cytology system, and evaluated its clinical feasibility for the management of malignant glioma. Materials and methods: Twenty-five selected patients with malignant glioma (newly diagnosed: 17; recurrent: 8) underwent surgical resection under photodiagnosis using photosensitizer Talaporfin sodium and a semiconductor laser. Intraoperatively, a crush smear preparation was made from a tiny amount of tumor tissue, and the fluorescence emitted upon 620/660 nm excitation was evaluated rapidly using a compact fluorescence microscope in the operating theater. Results: Fluorescence intensities of tumor tissues measured using a surgical microscope correlated with the tumor cell densities of tissues evaluated by measuring the red fluorescence emitted from the cytoplasm of tumor cells using a fluorescence microscope. A “weak fluorescence” indicated a reduction in the tumor cell density, whereas “no fluorescence” did not indicate the complete eradication of the tumor cells, but indicated that few tumor cells were emitting fluorescence.Conclusion: The rapid intraoperative detection of fluorescence from glioma cells using a compact fluorescence microscope was a useful to evaluate the presence of tumor cells in the resection cavity walls, and provides surgical implications for the more complete resection of malignant gliomas.

2021 ◽  
Vol 10 (22) ◽  
pp. 5375
Author(s):  
Jiro Akimoto ◽  
Shinjiro Fukami ◽  
Megumi Ichikawa ◽  
Kenta Nagai ◽  
Michihiro Kohno

Objective: The surgical eradication of malignant glioma cells is theoretically impossible. Therefore, reducing the number of remaining tumor cells around the brain–tumor interface (BTI) is crucial for achieving satisfactory clinical results. The usefulness of fluorescence–guided resection for the treatment of malignant glioma was recently reported, but the detection of infiltrating tumor cells in the BTI using a surgical microscope is not realistic. Therefore, we have developed an intraoperative rapid fluorescence cytology system, and exploratorily evaluated its clinical feasibility for the management of malignant glioma. Materials and methods: A total of 25 selected patients with malignant glioma (newly diagnosed: 17; recurrent: 8) underwent surgical resection under photodiagnosis using photosensitizer Talaporfin sodium and a semiconductor laser. Intraoperatively, a crush smear preparation was made from a tiny amount of tumor tissue, and the fluorescence emitted upon 620/660 nm excitation was evaluated rapidly using a compact fluorescence microscope in the operating theater. Results: Fluorescence intensities of tumor tissues measured using a surgical microscope correlated with the tumor cell densities of tissues evaluated by measuring the red fluorescence emitted from the cytoplasm of tumor cells using a fluorescence microscope. A “weak fluorescence” indicated a reduction in the tumor cell density, whereas “no fluorescence” did not indicate the complete eradication of the tumor cells, but indicated that few tumor cells were emitting fluorescence. Conclusion: The rapid intraoperative detection of fluorescence from glioma cells using a compact fluorescence microscope was probably useful to evaluate the presence of tumor cells in the resection cavity walls, and could provide surgical implications for the more complete resection of malignant gliomas.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi97-vi97
Author(s):  
Satoshi Suehiro ◽  
Takanori Ohnishi ◽  
Akihiro Inoue ◽  
Daisuke Yamashita ◽  
Masahiro Nishikawa ◽  
...  

Abstract OBJECTIVE High invasiveness of malignant gliomas frequently causes local tumor recurrence. To control such recurrence, novel therapies targeted toward infiltrating glioma cells are required. Here, we examined cytotoxic effects of sonodynamic therapy (SDT) combined with a sonosensitizer, 5-aminolevulinic acid (5-ALA), on malignant gliomas both in vitro and in vivo. METHODS In vitro cytotoxicity of 5-ALA-SDT was evaluated in U87 and U251 glioma cells and in U251Oct-3/4 glioma stemlike cells. Treatment-related apoptosis was analyzed using flow cytometry. Intracellular reactive oxygen species (ROS) were measured and the role of ROS in treatment-related cytotoxicity was examined. Effects of 5-ALA-SDT with high-intensity focused ultrasound (HIFU) on tumor growth, survival of glioma-transplanted mice, and histological features of the mouse brains were investigated. RESULTS The 5-ALA-SDT inhibited cell growth and changed cell morphology. Flow cytometric analysis indicated that 5-ALA-SDT induced apoptotic cell death. The 5-ALA-SDT generated higher ROS than in the control group, and inhibition of ROS generation completely eliminated the cytotoxic effects of 5-ALA-SDT. In the in vivo study, 5-ALA-SDT with HIFU greatly prolonged survival of the tumor-bearing mice compared with that of the control group (p < 0.05). Histologically, 5-ALA-SDT produced mainly necrosis of the tumor tissue in the focus area and induced apoptosis of the tumor cells in the perifocus area around the target of the HIFU-irradiated field. Normal brain tissues around the ultrasonic irradiation field of HIFU remained intact. CONCLUSIONS The 5-ALA-SDT was cytotoxic toward malignant gliomas. Generation of ROS by the SDT was thought to promote apoptosis of glioma cells. The 5-ALA-SDT with HIFU induced tumor necrosis in the focus area and apoptosis in the perifocus area of the HIFU-irradiated field. These results suggest that 5-ALA-SDT with HIFU may present a less invasive and tumor-specific therapy, not only for a tumor mass but also for infiltrating tumor cells in malignant gliomas.


2015 ◽  
Vol 3 (1) ◽  
pp. 57-61
Author(s):  
L. Lyubich ◽  
M. Lisyany

The use of neurogenic stem cells (NSCs) and neurogenic progenitor cells (NPCs) is one of the areas of brain and spinal cord lesions cell therapy. Intensive research of NSCs biology has revealed their tumor-tropic properties. Great migration potential and integration of NSCs in places of pathology in the central nervous system allows to consider their application as a means of targeted therapy of tumors. Antitumor properties of NSCs substantiate the development of treatment strategies for malignant gliomas using NSCs.The aim was to study the effect of rat neurogenic cells supernatant (NCS) on the tumor-inducing ability of glioma 101.8 cells at the intracerebral implantation in rats.Brain glioma 101.8 was modeling by intracerebral injection of 101.8-glioma cells suspension. NCS was received from whole rat brain tissue on 14th (E14) day of gestation.Modification of 101.8-glioma cells suspension by means of incubation with NCS (0.02 and 0.1 mg/ml) reduced the tumor-inducing ability of tumor cells, postponing the time of tumor clinical manifestations debut and increasing the lifetime of experimental animals.Under conditions of glioma induction with tumor cells, previously modified by NCS, cytotoxic activity of immune cells of tumor-bearing animals in MTT-test with allogeneic 101.8-glioma cells was increased.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e13026-e13026
Author(s):  
L. Keltner ◽  
J. C. Chen ◽  
S. Wang ◽  
M. L. Rakic

e13026 Background: Preclinical studies have shown that talaporfin sodium (LS11) can concentrate in brain-tumor cells and be activated by red light (664nm wavelength) from light-emitting diodes (LEDs). Singlet oxygen is released that induces blood-vessel occlusion and apoptosis in target tumor cells, which results in direct tumor-cell death and a potential antitumor immunogenic effect on untreated tumors. Preferential talaporfin uptake in tumor and clearance from normal surrounding brain occur by 24 hours after injection. A single center, open label phase IIa study was conducted to establish safety and acute antitumor effect of light-activated drug therapy in glioblastoma patients. Methods: Nine patients (Karnofsky score >90) with operable glioblastoma, four of which were recurrent in the right frontal or temporal lobe, were recruited at a neurosurgical center in Belgrade, Serbia. Tumor diameter ranged 4–9 cm. Treatment consisted of IV injection of 1mg/kg of LS11 with subsequent craniotomy after a 24-hour wait. A single LED device was inserted into the tumor center followed by different light doses, 250 joules in six patients and 500 joules in three patients. Treated tumors were then resected. Patients were assessed for 4 weeks with serial CT imaging, intracranial pressure monitoring, and clinical, neurologic, and lab examination. Results: No dose-limiting toxicity was noted at the higher light-dose level. Seven adverse events (2 neurologic) occurred in four patients, none treatment related. Apoptosis was evident by TUNEL stain, and vessel closure was seen histologically. No acute inflammatory changes or cerebral edema were noted; normal brain was unaffected. No adverse neurologic sequelae were ascribable to the therapy, and no post-operative ICP elevation was noted. At 4 weeks, all patients were alive; two patients progressed and died shortly thereafter. Conclusions: Treatment of glioblastoma with this light-activated drug therapy was safe and tolerable in this study. An antivascular effect and tumor-cell apoptosis were seen histologically and confined to tumor. [Table: see text]


2018 ◽  
Vol 51 (6) ◽  
pp. 2496-2508 ◽  
Author(s):  
Danfeng Zhang ◽  
Dawei Dai ◽  
Mengxia Zhou ◽  
Zhenxing Li ◽  
Chunhui Wang ◽  
...  

Background/Aims: Cyclin D1 (CCND1) is frequently overexpressed in malignant gliomas. We have previously shown ectopic overexpression of CCND1 in human malignant gliomas cell lines. Methods: Quantitative reverse transcriptase PCR (qRT-PCR) and Western Blot (WB) was performed to investigate the expression of CCND1 in glioma tissues and cell lines. The biological function of CCND1 was also investigated through knockdown and overexpression of BCYRN1 in vitro. Results: Here we reported that CCND1 expression was positively associated with the pathological grade and proliferative activity of astrocytomas, as the lowest expression was found in normal brain tissue (N = 3) whereas the highest expression was in high-grade glioma tissue (N = 25). Additionally, we found that the expression level of CCND1 was associated with IC50 values in malignant glioma cell lines. Forced inhibition of CCND1 increased temozolomide efficacy in U251 and SHG-44 cells. After CCND1 overexpression, the temozolomide efficacy decreased in U251 and SHG-44 cells. Colony survival assay and apoptosis analysis confirmed that CCND1 inhibition renders cells more sensitive to temozolomide treatment and temozolomide-induced apoptosis in U251 and SHG-44 cells. Inhibition of P-gp (MDR1) by Tariquidar overcomes the effects of CCND1 overexpression on inhibiting temozolomide-induced apoptosis. Inhibition of CCND1 inhibited cell growth in vitro and in vivo significantly more effectively after temozolomide treatments than single temozolomide treatments. Finally, inhibition of CCND1 in glioma cells reduced tumor volume in a murine model. Conclusion: Taken together, these data indicate that CCND1 overexpression upregulate P-gp and induces chemoresistance in human malignant gliomas cells and that inhibition of CCND1 may be an effective means of overcoming CCND1 associated chemoresistance in human malignant glioma cells.


2009 ◽  
Vol 111 (2) ◽  
pp. 230-237 ◽  
Author(s):  
Takeshi Miyazaki ◽  
Kouzo Moritake ◽  
Kazuo Yamada ◽  
Nobumasa Hara ◽  
Harumi Osago ◽  
...  

Object Indoleamine 2,3-dioxygenase (IDO), a kynurenine pathway (KP) enzyme catalyzing oxidation of the essential amino acid tryptophan (Trp), is thought to be involved in the immune resistance of malignant tumors through T-cell inactivation caused by Trp depletion and metabolite accumulation. Human malignant gliomas may use this strategy to escape immune attack. The object of this study was to investigate the possibility of IDO-dependent Trp depletion by malignant gliomas and the practicability of using an IDO inhibitor together with anticancer drugs to reserve Trp without decreasing the cytotoxicity of the drugs. Methods The authors studied expression of IDO and other KP enzymes and the effects of an IDO inhibitor, 1-methyl L-tryptophan (1MT), on Trp metabolism and cytotoxicity of anticancer drugs, together with direct measurement of KP metabolites, in cultured human malignant glioma cells. Results Upon interferon-γ (IFN-γ) stimulation, the glioma cells greatly increased their IDO mRNA expression concomitant with depletion of Trp. The IDO inhibitor 1MT successfully prevented Trp consumption by the stimulated glioma cells. Combining 1MT with anticancer drugs (temozolomide, bischloroethylnitrosourea [BCNU], etoposide and cisplatin) did not interfere with the drugs' suppression of growth of LN229 glioma cells but rather increased their inhibitory effects on IDO activity. Conclusions These findings suggest that the robust IDO expression with rapid consumption of Trp in human glioma cells induced by IFN-γ could lead to immune resistance in glioma cells. Indoleamine 2,3-dioxygenase inhibitors that prevent Trp depletion could be used with anticancer drugs to improve therapeutic effects.


2007 ◽  
Vol 107 (3) ◽  
pp. 617-627 ◽  
Author(s):  
Ilya V. Ulasov ◽  
Angel A. Rivera ◽  
Yu Han ◽  
David T. Curiel ◽  
Zeng B. Zhu ◽  
...  

Object Gene therapy protocols for malignant gliomas utilize adenoviral vectors that rely almost exclusively on the adenovirus serotype 5 (Ad5) backbone. The authors have previously shown that chimeric vectors that bind to the Ad3 receptor, or CD46, increase the transduction efficiency of malignant brain tumors. In light of the debate regarding the efficacy of CD46 compared with CD80/CD86 in binding Ad3 virions, the authors now examine the expression and transduction efficiency of Ad5/3 chimeras that bind via CD80/CD86. Methods The authors first analyzed CD80/CD86 expression in glioma cell lines. They then used three replication-defective vectors containing a luciferase reporter gene: Ad5/3 (containing the tail and shaft domain of Ad5 and the knob domain of Ad3); Ad3/5 (containing the tail of Ad5, shaft of Ad3, and knob of Ad5); and Ad3/3 (containing the tail of Ad5, shaft of Ad3, and knob of Ad3). These vectors were analyzed both in vitro and in vivo against malignant glioma cells. To examine further the effect of Ad5/3 fiber modification, the authors created an oncolytic vector, conditionally replicative Ad5/3 (CRAd5/3). Results The Ad5/3 vector showed a 10- to 100-fold enhanced transduction efficiency of malignant glioma compared with replication-defective wild-type adenovirus (reAd5) (p < 0.05). Moreover the use of Ad5/3 reduced transgene expression by more than 90% in normal human brain cells compared with reAd5. Finally, the use of CRAd5/3 inhibited tumor cell proliferation by 43% more than replication-competent wild-type virus in vitro (p < 0.05). Conclusions The results of this study demonstrate that the Ad5/3 vector offers superior transduction efficiency and low toxicity in the setting of brain tumors, and therefore represents a potential new approach to gene therapy for malignant gliomas.


2018 ◽  
Vol 129 (6) ◽  
pp. 1416-1428 ◽  
Author(s):  
Satoshi Suehiro ◽  
Takanori Ohnishi ◽  
Daisuke Yamashita ◽  
Shohei Kohno ◽  
Akihiro Inoue ◽  
...  

OBJECTIVEHigh invasiveness of malignant gliomas frequently causes early local recurrence of the tumor, resulting in extremely poor outcome. To control such recurrence, novel therapies targeted toward infiltrating glioma cells around the tumor border are required. Here, the authors investigated the antitumor activity of sonodynamic therapy (SDT) combined with a sonosensitizer, 5-aminolevulinic acid (5-ALA), on malignant gliomas to explore the possibility for clinical use of 5-ALA–mediated SDT (5-ALA-SDT).METHODSIn vitro cytotoxicity of 5-ALA-SDT was evaluated in U87 and U251 glioma cells and in U251Oct-3/4 glioma stemlike cells. Treatment-related apoptosis was analyzed using flow cytometry and TUNEL staining. Intracellular reactive oxygen species (ROS) were measured and the role of ROS in treatment-related cytotoxicity was examined by analysis of the effect of pretreatment with the radical scavenger edaravone. Effects of 5-ALA-SDT with high-intensity focused ultrasound (HIFU) on tumor growth, survival of glioma-transplanted mice, and histological features of the mouse brains were investigated.RESULTSThe 5-ALA-SDT inhibited cell growth and changed cell morphology, inducing cell shrinkage, vacuolization, and swelling. Flow cytometric analysis and TUNEL staining indicated that 5-ALA-SDT induced apoptotic cell death in all gliomas. The 5-ALA-SDT generated significantly higher ROS than in the control group, and inhibition of ROS generation by edaravone completely eliminated the cytotoxic effects of 5-ALA-SDT. In the in vivo study, 5-ALA-SDT with HIFU greatly prolonged survival of the tumor-bearing mice compared with that of the control group (p < 0.05). Histologically, 5-ALA-SDT produced mainly necrosis of the tumor tissue in the focus area and induced apoptosis of the tumor cells in the perifocus area around the target of the HIFU-irradiated field. The proliferative activity of the entire tumor was markedly decreased. Normal brain tissues around the ultrasonic irradiation field of HIFU remained intact.CONCLUSIONSThe 5-ALA-SDT was cytotoxic toward malignant gliomas. Generation of ROS by the SDT was thought to promote apoptosis of glioma cells. The 5-ALA-SDT with HIFU induced tumor necrosis in the focus area and apoptosis in the perifocus area of the HIFU-irradiated field, whereas the surrounding brain tissue remained normal, resulting in longer survival of the HIFU-treated mice compared with that of untreated mice. These results suggest that 5-ALA-SDT with HIFU may present a less invasive and tumor-specific therapy, not only for a tumor mass but also for infiltrating tumor cells in malignant gliomas.


2020 ◽  
Author(s):  
Ruifan Xie ◽  
Tobias Kessler ◽  
Julia Grosch ◽  
Ling Hai ◽  
Varun Venkataramani ◽  
...  

Abstract Background Malignant gliomas including glioblastomas are characterized by a striking cellular heterogeneity, which includes a subpopulation of glioma cells that becomes highly resistant by integration into tumor microtube (TM)-connected multicellular networks. Methods A novel functional approach to detect, isolate and characterize glioma cell subpopulations with respect to in vivo network integration is established, combining a dye staining method with intravital two-photon microscopy, FACS sorting, molecular profiling, and gene reporter studies. Results Glioblastoma cells that are part of the TM-connected tumor network show activated neurodevelopmental and glioma progression gene expression pathways. Importantly, many of them revealed profiles indicative of increased cellular stemness, including high expression of nestin. TM-connected glioblastoma cells also had a higher potential for re-initiation of brain tumor growth. Long-term tracking of tumor cell nestin expression in vivo revealed a stronger TM network integration and higher radioresistance of the nestin-high subpopulation. Glioblastoma cells that were both nestin-high and network-integrated were particularly able to adapt to radiotherapy with increased TM formation. Conclusion Multiple stem-like features are strongly enriched in a fraction of network-integrated glioma cells, explaining their particular resilience.


2019 ◽  
Vol 1 (Supplement_2) ◽  
pp. ii14-ii14
Author(s):  
Kazuhiko Kurozumi ◽  
Kentarou Fujii ◽  
Yosuke Shimazu ◽  
Yusuke Tomita ◽  
Yuji Matsumoto ◽  
...  

Abstract INTRODUCTION Malignant gliomas are one of the most common and aggressive intracranial neoplasms in humans. Expression of the gene encoding reduced expression in immortalized cells/Dickkopf-3 (REIC/Dkk-3) is reduced in a variety of human cancer cells. We previously showed the antitumor effect of an adenoviral vector carrying REIC/Dkk-3 gene (Ad-CAG-REIC). Recently, we have also developed a novel adenoviral vector expressing REIC/Dkk-3 (Ad-SGE-REIC). We assessed the anti-glioma effect of the Ad-SGE-REIC and planned a clinical trial of Ad-SGE-REIC for malignant glioma. MATERIALS AND METHODS We evaluated a cytotoxicity assay to treatments with Ad-SGE-REIC, Ad-CAG-REIC, or Ad-LacZ (control) using malignant glioma cells. The survival of mice in each group was analyzed by the Kaplan-Meier method. We also performed Good Laboratory Practice (GLP) toxicology tests and prepared a protocol for this clinical trial. RESULTS The treatment with Ad-SGE-REIC showed the number of malignant glioma cells attached to the bottom of culture wells was significantly reduced in a time-dependent manner. Mice treated with Ad-SGE-REIC significantly prolonged survival time more than those treated with other vectors. A cGMP product of Ad-SGE-REIC was developed and supplied by a startup biotech company, Momotaro-Gene Inc. We conducted GLP toxicology tests using the intracranial injection of higher doses of Ad-SGE-REIC at Shin Nippon Biomedical Laboratories (SNBL Japan). After finishing the consultation with Pharmaceuticals and Medical Devices Agency (PMDA), we prepared a protocol for a phase I/IIa clinical trial of Ad-SGE-REIC for the treatment of recurrent malignant glioma with our academic research organization (ARO), supported by Japan Agency for Medical Research and Development (AMED). This protocol was reviewed by our institution review board in March 2019. We submitted a notification of this trial in April 2019. CONCLUSIONS We demonstrated the anti-glioma effect of Ad-SGE-REIC. We start a phase I/IIa clinical trial of Ad-SGE-REIC for the treatment of recurrent malignant glioma (https://jrct.niph.go.jp/en-latest-detail/jRCT2063190013).


Sign in / Sign up

Export Citation Format

Share Document