1.The role of interleukin-18 in glioblastoma pathology implies therapeutic potential of two old drugs-disulfiram and ritonavir.
Chinese Journal of Cancer 2015;34(4):161-165
Based on reporting in the last several years, an impressive but dismal list of cytotoxic chemotherapies that fail to prolong the median overall survival of patients with glioblastoma has prompted the development of treatment protocols designed to interfere with growth-facilitating signaling systems by using non-cytotoxic, non-oncology drugs. Recent recognition of the pro-mobility stimulus, interleukin-18, as a driver of centrifugal glioblastoma cell migration allows potential treatment adjuncts with disulfiram and ritonavir. Disulfiram and ritonavir are well-tolerated, non-cytotoxic, non-oncology chemotherapeutic drugs that are marketed for the treatment of alcoholism and human immunodeficiency virus (HIV) infection, respectively. Both drugs exhibit an interleukin-18-inhibiting function. Given the favorable tolerability profile of disulfiram and ritonavir, the unlikely drug-drug interaction with temozolomide, and the poor prognosis of glioblastoma, trials of addition of disulfiram and ritonavir to current standard initial treatment of glioblastoma would be warranted.
Antineoplastic Agents
;
Dacarbazine
;
analogs & derivatives
;
Disulfiram
;
Glioblastoma
;
Humans
;
Interleukin-18
;
Ritonavir
2.Caspase-independent programmed cell death induced by temozolomide in rat glioma C6 cell line.
Journal of Southern Medical University 2015;35(2):229-233
OBJECTIVETo investigate programmed cell death induced by temozolomide in rat glioma C6 cell line.
METHODSRat glioma C6 cell line was treated by temozolomide at different concentrations and for different time lengths. MTT assay was used to evaluate the cell inhibition rate to determine the optimal exposure time and concentration. After the exposure to 400 µg/ml temozolomide for 24 h, the cells were observed for programmed cell death using HE staining, Hochest and Tunnel assay, Western blotting, and immunohistochemistry.
RESULTSMTT, HE staining, and Hochest and Tunnel assay all showed temozolomide-induced apoptosis in rat glioma C6 cell line. Western blotting and immunohistochemistry revealed up-regulation of Bax and down-regulation of Bcl-2 expression in the exposed cells, where the expressions of caspases 3, 8, 9, and 12 remained unchanged.
CONCLUSIONTemozolomide induces apoptosis in rat glioma C6 cell line through a caspase- independent pathway.
Animals ; Apoptosis ; Caspases ; Cell Line, Tumor ; Dacarbazine ; analogs & derivatives ; pharmacology ; Down-Regulation ; Glioma ; pathology ; Rats ; Up-Regulation
3.RITA combined with temozolomide inhibits the proliferation of human glioblastoma U87 cells.
Xiao-Yan HE ; Xiao-Li FENG ; Xin-Pei SONG ; Huan-Chao ZENG ; Zhong-Xu CAO ; Wei-Wei XIAO ; Bao ZHANG ; Qing-Hua WU
Journal of Southern Medical University 2016;36(10):1423-1428
OBJECTIVETo observe the effect of RITA, a small molecule that targets p53, combined with temozolomide (TMZ) on proliferation, colony formation and apoptosis of human glioblastoma U87 cells and explore the underlying mechanism.
METHODSCultured U87 cells were treated with RITA (1, 5, 10, 20 µmol/L), TMZ, or RITA+TMZ (half dose) for 24, 48 or 72 h. MTS assay were used to detect the cell proliferation, and the cell proliferation rate and inhibitory rate were calculated. The effect of combined treatments was evaluated by the q value. The expressions of p53, p21 and other apoptosis-associated genes were detected by qRT-PCR and Western blotting; cell apoptosis was assayed using flow cytometry with Annexin V/PI double staining; colony formation of the cells was detected with crystal violet staining.
RESULTSMTS assay showed that RITA at the 4 doses more potently inhibited U87 cell viability than TMZ at 72 h (P=0.000) with inhibitory rates of 25.94%-41.38% and 3.84%-8.20%, respectively. RITA combined with TMZ caused a more significant inhibition of U87 cells (29.21%-52.11%) than RITA (P<0.01) and TMZ (P=0.000) alone. At the doses above 5 µmol/L, the combined treatments with RITA+TMZ for 48 h resulted in q values exceeding 1.2 and showed an obvious synergistic effect of the drugs. Both RITA and TMZ, especially the latter, significantly increased the expressions of p53, p21, puma, and other apoptosis-associated genes to accelerate apoptosis and inhibit the growth and colony formation of U87 cells, and the effect was more obvious with a combined treatment.
CONCLUSIONRITA inhibits the growth of human glioblastoma cells and enhance their sensitivity to TMZ by up-regulating p53 expression, and when combined, RITA and TMZ show a synergistic effect to cause a stronger cell inhibition.
Apoptosis ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; Cell Survival ; Dacarbazine ; analogs & derivatives ; pharmacology ; Furans ; pharmacology ; Glioblastoma ; drug therapy ; Humans
4.Orthogonal experiments for optimizing the formulation and preparation conditions of temozolomide solid lipid nanoparticles.
Mingjin DOU ; Guihua HUANG ; Yanwei XI ; Na ZHANG
Journal of Biomedical Engineering 2008;25(5):1141-1145
TMZ-SLN were prepared by emulsification-low temperature solidification method with stearic acid. The formulation and the preparation conditions were optimized by orthogonal experiments using entrapment efficiency as the evaluation index. The morphology was detected by transmission electron microscope. The Zeta potentials and the particle size distribution were evaluated by Laser Doppler Anemometry. The entrapment efficiencies and the drug release characteristics in vitro were assessed. The result showed that TMZ-SLN were concinnous and spherical in shape. The mean diameter (d(av) ) was 65.0 +/- 6.2 nm and the Zeta potential was -37.2 mV. The average entrapment efficiency was 58.9% +/- 1.21 %. The drug release behavior in vitro conformed to Higuchi Equation. The formation of a new material phase was testified by analysis of differential scanning calorimetry.
Antineoplastic Agents, Alkylating
;
administration & dosage
;
chemistry
;
Dacarbazine
;
administration & dosage
;
analogs & derivatives
;
chemistry
;
Drug Carriers
;
chemistry
;
Lipids
;
chemistry
;
Nanoparticles
;
chemistry
;
Particle Size
;
Stearic Acids
;
chemistry
5.Does Early Postsurgical Temozolomide Plus Concomitant Radiochemotherapy Regimen Have Any Benefit in Newly-diagnosed Glioblastoma Patients? A Multi-center, Randomized, Parallel, Open-label, Phase II Clinical Trial.
Ying MAO ; Yu YAO ; Li-Wei ZHANG ; Yi-Cheng LU ; Zhong-Ping CHEN ; Jian-Min ZHANG ; Song-Tao QI ; Chao YOU ; Ren-Zhi WANG ; Shu-Yuan YANG ; Xiang ZHANG ; Ji-Sheng WANG ; Ju-Xiang CHEN ; Qun-Ying YANG ; Hong SHEN ; Zhi-Yong LI ; Xiang WANG ; Wen-Bin MA ; Xue-Jun YANG ; Hai-Ning ZHEN ; Liang-Fu ZHOU
Chinese Medical Journal 2015;128(20):2751-2758
BACKGROUNDThe radiochemotherapy regimen concomitantly employing temozolomide (TMZ) chemotherapy and radiotherapy (RT) 4 weeks after surgery, followed by 6 cycles of TMZ is a common treatment for glioblastoma (GBM). However, its median overall survival (OS) is only 14.6 months. This study was to explore the effectiveness and safety of early TMZ chemotherapy between surgery and chemoradiotherapy plus the standard concomitant radiochemotherapy regimen.
METHODSA randomized, parallel group, open-label study of 99 newly diagnosed GBM patients was conducted at 10 independent Chinese neurosurgical departments from June 2008 to June 2012. Patients were treated with concomitant radiochemotherapy regimen plus early postsurgical temozolomide (early TMZ group) or standard concomitant radiochemotherapy regimen (control group). Overall response was assessed based on objective tumor assessments, administration of corticosteroid and neurological status test. Hematological, biochemical, laboratory, adverse event (AE), and neurological condition were measured for 24 months of follow-up. The primary efficacy endpoint of this study was overall survival (OS). The secondary endpoint was progression free survival (PFS).
RESULTSThe median OS time in the early TMZ group was 17.6 months, compared with 13.2 months in the control group (log-rank test P = 0.021). In addition, the OS rate in the early TMZ group was higher at 6, 12, and 18 months than in the control group, respectively (P < 0.05). The median PFS time was 8.7 months in the early TMZ group and 10.4 months in the control group (log-rank test P = 0.695). AEs occurred in 29 (55.8%) and 31(73.8%) patients respectively in early and control groups, including nausea (15.4% vs. 33.3%), vomiting (7.7% vs. 28.6%), fever (7.7% vs. 11.9%), and headache (3.8% vs. 23.8%). Only 30.8% and 33.3% were drug-related, respectively.
CONCLUSIONSAddition of TMZ chemotherapy in the early break of the standard concomitant radiochemotherapy regimen was well tolerated and significantly improved the OS of the GBM patients, compared with standard concomitant radiochemotherapy regimen. However, a larger randomized trial is warranted to verify these results.
Adult ; Aged ; Antineoplastic Agents, Alkylating ; therapeutic use ; Chemoradiotherapy ; methods ; Dacarbazine ; analogs & derivatives ; therapeutic use ; Glioblastoma ; drug therapy ; radiotherapy ; Humans ; Middle Aged ; Treatment Outcome ; Young Adult
6.Mechanism of thalidomide to enhance cytotoxicity of temozolomide in U251-MG glioma cells in vitro.
Song GAO ; Xue-jun YANG ; Wen-gao ZHANG ; Yan-wei JI ; Qiang PAN
Chinese Medical Journal 2009;122(11):1260-1266
BACKGROUNDGlioma is the most common primary brain tumor with poor prognosis. Temozolomide has been used with thalidomide to treat gliomas. We investigated the synergistic mechanism of these two drugs in vitro.
METHODSHuman malignant glioma cells U251-MG were cultured and assigned to four groups with different treatments for 3 days: temozolomide group (100 micromol/L), thalidomide group (100 microg/L), temozolomide (100 micromol/L) plus thalidomide group (100 microg/L) and control group. MTT assay was applied to evaluate the cell viability. Cell cycle was analyzed by flow cytometry. The ultra-structural features of autophagosomes were observed with electron microscope. Acridine orange and monodansylcadaverine were adopted to label autophagosomes and flow cytometry was applied for quantification of autophagosomes. The expression of autophagy-associated protein was detected by Western blotting.
RESULTSProliferation of tumor cell was obviously suppressed by temozolomide with thalidomide treatment than by either drug used alone (P = 0.000 for each day). The combination treatment induced cell cycle arrest at G0/G1 phase. Typical autophagic ultra-structural character was found after the combined treatment. Thalidomide promoted the autophagy induced by temozolomide. The autophagy-associated proteins-microtubule associated protein 1 light chain 3 (MAP1LC3) and Beclin1 were more significantly up-regulated by the combined treatment than temozolomide used alone (MAP1LC3, P = 0.000; Beclin1, P = 0.004). The expression level of phosphatase and tensin homolog deleted on chromosome ten (PTEN), which promoted autophagy by suppressing PI3K/Akt/mTOR signaling pathway, was elevated by thalidomide (thalidomide group: P = 0.000; combined group: P = 0.002).
CONCLUSIONSThalidomide enhances the cytotoxicity of temozolomide by promoting the autophagy induced by temozolomide. Contributing to the up-regulation of PTEN by thalidomide, the expression of autophagy associated protein-MAP1LC3 and Beclin1 was enhanced, which leads to a reinforced autophagy in the combined treatment of temozolomide and thalidomide in vitro.
Antineoplastic Agents, Alkylating ; pharmacology ; Autophagy ; drug effects ; Cell Line, Tumor ; Cell Proliferation ; drug effects ; Dacarbazine ; analogs & derivatives ; pharmacology ; Glioma ; pathology ; Humans ; Thalidomide ; pharmacology
7.Opposite Interplay Between the Canonical WNT/β-Catenin Pathway and PPAR Gamma: A Potential Therapeutic Target in Gliomas.
Alexandre VALLÉE ; Yves LECARPENTIER ; Rémy GUILLEVIN ; Jean-Noël VALLÉE
Neuroscience Bulletin 2018;34(3):573-588
In gliomas, the canonical Wingless/Int (WNT)/β-catenin pathway is increased while peroxisome proliferator-activated receptor gamma (PPAR-γ) is downregulated. The two systems act in an opposite manner. This review focuses on the interplay between WNT/β-catenin signaling and PPAR-γ and their metabolic implications as potential therapeutic target in gliomas. Activation of the WNT/β-catenin pathway stimulates the transcription of genes involved in proliferation, invasion, nucleotide synthesis, tumor growth, and angiogenesis. Activation of PPAR-γ agonists inhibits various signaling pathways such as the JAK/STAT, WNT/β-catenin, and PI3K/Akt pathways, which reduces tumor growth, cell proliferation, cell invasiveness, and angiogenesis. Nonsteroidal anti-inflammatory drugs, curcumin, antipsychotic drugs, adiponectin, and sulforaphane downregulate the WNT/β-catenin pathway through the upregulation of PPAR-γ and thus appear to provide an interesting therapeutic approach for gliomas. Temozolomide (TMZ) is an antiangiogenic agent. The downstream action of this opposite interplay may explain the TMZ-resistance often reported in gliomas.
Animals
;
Brain Neoplasms
;
metabolism
;
therapy
;
Dacarbazine
;
analogs & derivatives
;
pharmacology
;
Down-Regulation
;
drug effects
;
Glioma
;
metabolism
;
therapy
;
Humans
;
PPAR gamma
;
metabolism
;
Temozolomide
;
Wnt Signaling Pathway
;
drug effects
;
physiology
8.Combination Therapy with Rituximab and Temozolomide for Recurrent and Refractory Primary Central Nervous System Lymphoma.
Mineko MURAKAMI ; Takamitsu FUJIMAKI ; Shuichiro ASANO ; Hiroshi NAKAGUCHI ; Shoko M YAMADA ; Katsumi HOYA ; Kazuto YAMAZAKI ; Yasuo ISHIDA ; Akira MATSUNO
Yonsei Medical Journal 2011;52(6):1031-1034
High-dose methotrexate-based chemotherapy has extended survival in patients with primary central nervous system lymphoma (PCNSL). However, although salvage treatment is necessary in recurrent and refractory PCNSL, this has not been standardized. We herein describe the efficacy of a combination of rituximab and temozolomide (TMZ) in two consecutive patients with recurrent and refractory PCNSL. Based on the immunohistochemical study, case 1 had a non-germinal center B-cell-like (non-GCB) subtype, was positive for bcl-2 and negative for O6-methylguanine-DNA methyltransferase (MGMT). Case 2 was GCB subtype, bcl-2-, and MGMT+. Because of the positive expression of MGMT, interferon-beta was additionally given in case 2. Complete responses and partial responses were obtained after the third and fourth cycles of combination therapy, respectively. This was maintained for 12 months, with acceptable toxicity. The combination of rituximab and TMZ was effective in tumors with different immunohistochemical profiles. This combination therapy warrants further study in a larger population.
Aged
;
Antibodies, Monoclonal, Murine-Derived/*therapeutic use
;
Antineoplastic Agents/*therapeutic use
;
Central Nervous System Neoplasms/*drug therapy
;
Dacarbazine/*analogs & derivatives/therapeutic use
;
Drug Therapy, Combination/*methods
;
Humans
;
Lymphoma/*drug therapy
;
Male
;
Middle Aged
;
Neoplasm Recurrence, Local/drug therapy
9.Intranasal administration of temozolomide for brain-targeting delivery: therapeutic effect on glioma in rats.
Ying LI ; Yongliang GAO ; Gang LIU ; Xiudi ZHOU ; Yan WANG ; Yulin WANG ; Lin MA
Journal of Southern Medical University 2014;34(5):631-635
OBJECTIVETo study the therapeutic effect of intranasal administration of temozolomide (TMZ) for brain-targeting delivery in a rat model bearing orthotopic C6 glioma xenografts.
METHODSForty Wistar rat bearing brain C6 glioma xenograft were randomly divided into 4 groups and treated with physiological saline solution or with TMZ by intravenous injection, gavage or intranasal administration. The tumor size, rat survival time and pathological changes were observed in each group.
RESULTSMagnetic resonance imaging showed a significantly reduced volume of glioma in intranasal TMZ group compared with that in the control, intraveneous TMZ injection group and TMZ gavage groups (12.45∓2.49 mm(3) vs 60.16∓4.12, 33.17∓3.56, and 35.16∓4.36 mm(3), respectively, P<0.05). The median survival time of the C6 glioma-bearing rats was also significantly longer in intranasal TMZ group than in the other 3 groups (31.0 days vs 20, 19, and 21.5 days, respectively, P<0.05). In the glioma xenografts, PCNA expression was the lowest and tumor cell apoptosis rate the highest in intranasal TMZ group.
CONCLUSIONIntranasal TMZ administration can suppress the growth of C6 glioma in rats and may serve as an effective strategy for glioma treatment.
Administration, Intranasal ; Animals ; Antineoplastic Agents, Alkylating ; administration & dosage ; Apoptosis ; Brain Neoplasms ; drug therapy ; Cell Line, Tumor ; Dacarbazine ; administration & dosage ; analogs & derivatives ; Drug Delivery Systems ; Glioma ; drug therapy ; Magnetic Resonance Imaging ; Neoplasm Transplantation ; Rats ; Rats, Wistar
10.Histone deacetylase inhibitor, 2-propylpentanoic acid, increases the chemosensitivity and radiosensitivity of human glioma cell lines in vitro.
Cui-jie SHAO ; Ming-wei WU ; Fu-rong CHEN ; Cong LI ; Yun-fei XIA ; Zhong-ping CHEN
Chinese Medical Journal 2012;125(24):4338-4343
BACKGROUNDTreatment for malignant glioma generally consists of cytoreductive surgery followed by radiotherapy and chemotherapy. In this study, we intended to investigate the effects of 2-propylpentanoic acid (VPA), a histone deacetylase inhibitor, on chemosensitivity and radiosensitivity in human glioma cell lines.
METHODSHuman glioma cell lines, T98-G, and SF295, were treated with temozolomide (TMZ) or irradiation (IR), with or without VPA (1.0 mmol/L). Then, cytotoxicity and clonogenic survival assay was performed. Cell cycle stage, apoptosis, and autophagy were also detected using flow cytometry and dansyl monocadaverin (MDC) incorporation assay. One-way analysis of variance (ANOVA) and t-test were used to analyze the differences among variant groups.
RESULTSMild cytotoxicity of VPA was revealed in both cell lines, T98-G and SF295, with the 50% inhibiting concentration (IC50) value of (3.85 ± 0.58) mmol/L and (2.15 ± 0.38) mmol/L, respectively; while the IC50 value of TMZ was (0.20 ± 0.09) mmol/L for T98-G and (0.08 ± 0.02) mmol/L for SF295. Moreover, if combined with VPA (1.0 mmol/L) for 96 hours, the sensitivity of glioma cells to TMZ was significant increased (P < 0.05). The surviving fractions at 2 Gy (SF2) of T98-G and SF295 cells exposed to IR alone were 0.52 and 0.58. However, when VPA was combined with IR, the SF2 of T98-G and SF295 dropped to 0.39 (P = 0.047) and 0.49 (P = 0.049), respectively. Treatment with VPA plus TMZ or IR also resulted in a significant decrease in the proportion of cells in the G2 phase and increased apoptotic rates as well as autophagy in T98-G and SF295 cell lines (P < 0.01).
CONCLUSIONVPA may enhance the activities of TMZ and IR on glioma cells possibly through cell cycle block and promote autophagy, and thus could be a potential sensitizer of glioma treatment.
Apoptosis ; drug effects ; radiation effects ; Blotting, Western ; Cell Line, Tumor ; Cell Survival ; drug effects ; radiation effects ; Dacarbazine ; analogs & derivatives ; pharmacology ; Flow Cytometry ; Glioma ; metabolism ; Histone Deacetylase Inhibitors ; pharmacology ; Humans ; Valproic Acid ; pharmacology