1
|
Bai X, Xing H, Feng M, Ma W, Wang S. Dose and Efficacy of Bevacizumab in Recurrent High-Grade Gliomas: A Retrospective Study. Cancer Manag Res 2024; 16:1617-1626. [PMID: 39575164 PMCID: PMC11578801 DOI: 10.2147/cmar.s481289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024] Open
Abstract
Purpose We retrospectively analyzed the effect of Bevacizumab (BEV) on recurrent high-grade glioma (rHGG) and examined the relationship between dose and efficacy. Methods A total of 182 patients with rHGG were included in this study. Patients were divided into a non-BEV group and a BEV group according to the treatment they received, and the BEV group was further divided into a low-dose group and a high-dose group based on the dose. Depending on the number of groups and the characteristics of numerical variables, t-test, ANOVA, or rank-sum test were selected. Categorical variables were compared using the chi-squared test. Results Progression-free survival (PFS) was lower in the non-BEV group compared to the BEV group, while overall survival (OS) was not different between the two groups. There was no difference in PFS and OS between low-dose group and high-dose group. Notably, we found that patients with longer PFS and OS were more likely to be from the BEV group. In addition, differences in Karnofsky Performance Score (KPS), steroid dose, and brain edema were observed in the non-BEV, low-dose, and high-dose groups from 3 to 12 months after treatment. Conclusion BEV can improve PFS in patients with rHGG, although its impact on OS is limited. There was no difference in the efficacy of different doses of BEV on rHGG. Interestingly, patients with longer PFS and OS were more likely to be from the BEV group. Based on these findings, long-term low-dose BEV appears to be an effective treatment option for rHGG.
Collapse
Affiliation(s)
- Xuexue Bai
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hao Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Ming Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Shiyong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
2
|
Walter T, Foray G, Mohammed-Brahim N, Levé C, Mandonnet E, Gayat E. Mini-strokes after awake surgery for glioma resection: are there anesthesia related factors? Acta Neurochir (Wien) 2024; 166:310. [PMID: 39085454 DOI: 10.1007/s00701-024-06195-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/06/2024] [Indexed: 08/02/2024]
Abstract
INTRODUCTION Awake surgery is now a common approach for the resection of glioma. One of the surgical complications is mini-stroke which take the form of periresectional small areas of brain ischemic lesions. The main objective of this study is to evaluate the association between factors related to anesthetic management and the risk of mini-stroke, in awake surgery for glioma resection. METHODS In this single-center retrospective study, all patients who were operated on, between 2011 and 2022, in awake conditions for a glioma resection, were retrospectively included. The studied anesthetic parameters included hemodynamic variables, fluid intake and urinary output. The primary endpoint was the presence of mini-stroke on a magnetic resonance imaging performed within the first 48 h postoperatively. RESULTS A total of 176 surgeries were included. Mini-stroke was present in 120/171 surgeries (70%), with a median volume of 1.2 interquartile range [0.4-2.2] cubic centimeters (cc). In a multivariable analysis, only the per operative urinary output was significantly associated with the incidence of postoperative mini-strokes (adjusted odd-ratio 0.65, 95% confidence interval 0.45-0.94, p = 0.02). No variables related to the anesthetic management were associated with the volume of postoperative mini-strokes. In particular, the time spent below 90% of the baseline systolic blood pressure was not associated with either the risk or the volume of mini-strokes. CONCLUSION During awake surgery for glioma resection, among several anesthesia related factors, only the per operative urinary output was associated with the incidence of postoperative mini-stroke.
Collapse
Affiliation(s)
- Thaïs Walter
- Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Lariboisière Hospitals, FHU PROMICE, DMU Parabol, Paris, France.
- Université Paris Cité, Paris, France.
- UMR-S 942 (MASCOT), INSERM, Paris, France.
| | - Grégoire Foray
- Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Lariboisière Hospitals, FHU PROMICE, DMU Parabol, Paris, France
| | | | - Charlotte Levé
- Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Lariboisière Hospitals, FHU PROMICE, DMU Parabol, Paris, France
| | - Emmanuel Mandonnet
- Service of Neurosurgery, Lariboisière Hospital, AP/HP Nord, Paris, France
- Université Paris Cité, Paris, France
| | - Etienne Gayat
- Department of Anesthesiology and Critical Care and Burn Unit, Saint-Louis and Lariboisière Hospitals, FHU PROMICE, DMU Parabol, Paris, France
- Université Paris Cité, Paris, France
- UMR-S 942 (MASCOT), INSERM, Paris, France
| |
Collapse
|
3
|
Barbosa LC, Machado GC, Heringer M, Ferrer VP. Identification of established and novel extracellular matrix components in glioblastoma as targets for angiogenesis and prognosis. Neurogenetics 2024; 25:249-262. [PMID: 38775886 DOI: 10.1007/s10048-024-00763-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/10/2024] [Indexed: 07/16/2024]
Abstract
Glioblastomas (GBM) are aggressive tumors known for their heterogeneity, rapid proliferation, treatment resistance, and extensive vasculature. Angiogenesis, the formation of new vessels, involves endothelial cell (EC) migration and proliferation. Various extracellular matrix (ECM) molecules regulate EC survival, migration, and proliferation. Culturing human brain EC (HBMEC) on GBM-derived ECM revealed a decrease in EC numbers compared to controls. Through in silico analysis, we explored ECM gene expression differences between GBM and brain normal glia cells and the impact of GBM microenvironment on EC ECM transcripts. ECM molecules such as collagen alpha chains (COL4A1, COL4A2, p < 0.0001); laminin alpha (LAMA4), beta (LAMB2), and gamma (LAMC1) chains (p < 0.0005); neurocan (NCAN), brevican (BCAN) and versican (VCAN) (p < 0.0005); hyaluronan synthase (HAS) 2 and metalloprotease (MMP) 2 (p < 0.005); MMP inhibitors (TIMP1-4, p < 0.0005), transforming growth factor beta-1 (TGFB1) and integrin alpha (ITGA3/5) (p < 0.05) and beta (ITGB1, p < 0.0005) chains showed increased expression in GBM. Additionally, GBM-influenced EC exhibited elevated expression of COL5A3, COL6A1, COL22A1 and COL27A1 (p < 0.01); LAMA1, LAMB1 (p < 0.001); fibulins (FBLN1/2, p < 0.01); MMP9, HAS1, ITGA3, TGFB1, and wingless-related integration site 9B (WNT9B) (p < 0.01) compared to normal EC. Some of these molecules: COL5A1/3, COL6A1, COL22/27A1, FBLN1/2, ITGA3/5, ITGB1 and LAMA1/B1 (p < 0.01); NCAN, HAS1, MMP2/9, TIMP1/2 and TGFB1 (p < 0.05) correlated with GBM patient survival. In conclusion, this study identified both established and novel ECM molecules regulating GBM angiogenesis, suggesting NCAN and COL27A1 are new potential prognostic biomarkers for GBM.
Collapse
Affiliation(s)
- Lucas Cunha Barbosa
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil
| | - Gabriel Cardoso Machado
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil
| | - Manoela Heringer
- Brain's Biomedicine Lab, Paulo Niemeyer State Brain Institute, Rio de Janeiro, Brazil
| | - Valéria Pereira Ferrer
- Graduation Program of Pathological Anatomy, Faculty of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Laboratory of Cellular and Molecular Biology of Tumors, Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niteroi, Brazil.
| |
Collapse
|
4
|
Khathayer F, Mikael M. Mocetinostat as a novel selective histone deacetylase (HDAC) inhibitor in the promotion of apoptosis in glioblastoma cell line C6 and T98G. RESEARCH SQUARE 2024:rs.3.rs-4170668. [PMID: 38645087 PMCID: PMC11030514 DOI: 10.21203/rs.3.rs-4170668/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Histon deacetylase (HDAC) enzyme is one of the enzymes involved in regulating gene expression and epigenetic alternation of cells by removing acetyl groups from lysine residue on a histone, allowing the histones to wrap the DNA more tightly and suppressing a tumor-suppressing gene. HDAC inhibitors play an important role in inhibiting the proliferation of tumor cells by restricting the mechanism of action of HDAC enzyme, leading to the addition of acetyl groups to lysine. Mocetinostat, also known by its chemical name (MGCD0103), is a novel isotype selective HDAC enzyme that explicitly targets HDAC isoforms inhibiting Class1(HDAC 1,2,3,8) and Class IV (HDAC11) enzymes. It was approved for treating the phase II trial of Hodgkin's lymphoma in 2010. Our study revealed that different doses of Mocetinostat inhibit the growth of glioblastoma cells, metastasis, and angiogenesis and induce the apoptosis and differentiation of glioblastoma cells C6 and T98G. Western blot has shown that MGCD0103 has many biological activities to control glioblastoma cancer cells. MGCD0103 can modulate the molecular mechanism for several pathways in cells, such as inhibition of the PI3K/AKT pathway and suppression of HDAC1 enzyme activity in charge of many biological processes in the initiation and progression of cancer. The high doses of Mocetinostat drug significantly induce apoptosis and suppress cancer cell proliferation through increased pro-apoptotic proteins (BAX) and a down level of anti-apoptotic proteins(Bid, Bcl2). Also, the mocetinostat upregulated the expression of the tumor suppressor gene and downregulated the gene expression of the E2f1 transcription factor. Additionally, MGCDO103-induced differentiation was facilitated by activating the differentiation marker GFAP and preventing the undifferentiation marker from expression (Id2, N-Myc). The MGCD0103 is a potent anticancer drug crucial in treating glioblastoma cells.
Collapse
|
5
|
Nikitin PV, Musina GR, Polozov VN, Goreiko DN, Krasnovsky VM, Werkenbark L, Kjelin M, Timashev PS. Development of Glioblastoma from Stem Cells to a Full-Fledged Tumor. Turk Patoloji Derg 2023; 39:117-132. [PMID: 35876685 PMCID: PMC10518198 DOI: 10.5146/tjpath.2022.01582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE IDH wild-type glioblastomas (GBM) are one of the most malignant and complex tumors for treatment. The urgent question of new therapeutic and diagnostic tools searching should be resolved based on cellular and molecular pathogenesis mechanisms, which remain insufficiently studied. In this study, we aimed to investigate GBM pathogenesis. MATERIAL AND METHOD /b > Using the isolation of different GBM cell populations and the cell cultures, animal models, and molecular genetic methods, we tried to clarify the picture of GBM pathogenesis by constructing a projection from different glioma stem cells types to an integral neoplasm. RESULTS We have shown a potential transformation pathway for both glioma stem cells and four definitive cell populations during gliomagenesis. Moreover, we have characterized each population, taking into account its place in the pathogenetic continuum, with a description of the most fundamental molecular and functional properties. CONCLUSION Finally, we have formed a complex holistic concept of the pathogenetic evolution of GBM at the cell-population level by integrating our results with the data of the world literature.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Piotr Sergeevich Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia; Chemistry Department, Lomonosov Moscow State University, Moscow, Russia; World-Class Research Center “Digital biodesign and personalized healthcare,” Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
6
|
Eidizade F, Soukhtanloo M, Zhiani R, Mehrzad J, Mirzavi F. Inhibition of glioblastoma proliferation, invasion, and migration by Urolithin B through inducing G0/G1 arrest and targeting MMP-2/-9 expression and activity. Biofactors 2022; 49:379-389. [PMID: 36310375 DOI: 10.1002/biof.1915] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
One kind of brain cancer with a dismal prognosis is called glioblastoma multiforme (GBM) due to its high growth rate and widespread tumor cell invasion into various areas of the brain. To improve therapeutic approaches, the objective of this research investigates the cytotoxic, anti-metastatic, and apoptotic effect of urolithin-B (UB) as a bioactive metabolite of ellagitannins (ETs) on GBM U87 cells. The malignant GBM cell line (U87) was examined for apoptosis rate, cell cycle analysis, cell viability, mRNA expressions of several apoptotic and metastasis-associated genes, production of reactive oxygen species (ROS), MMP-2, and MMP-9 activity and protein expression, and migration ability. The findings revealed that UB decreased U87 GBM viability in a dose-dependent manner and NIH/3T3 normal cells with the IC50 value of 30 and 55 μM after 24 h, respectively. UB also induces necrosis and G0/G1 cell cycle arrest in U87 cells. UB also increases ROS production and caused down-regulation of Bcl2 and up-regulation of Bax apoptotic genes. Additionally, treatment of UB reduced the migration of U87 cells. The protein levels, mRNA expression, and the MMP-2 and MMP-9 enzyme activities also decreased concentration-dependently. So, due to the non-toxic nature of UB and its ability to induce apoptosis and reduce the U87 GBM cell invasion and migration, after more research, it can be regarded as a promising new anti-GBM compound.
Collapse
Affiliation(s)
- Fateme Eidizade
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mohammad Soukhtanloo
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmacological Research Center of Medicinal Plants, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rahele Zhiani
- Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
- New Materials Technology and Processing Research Center, Department of Chemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Jamshid Mehrzad
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
7
|
Zaman N, Dass SS, DU Parcq P, Macmahon S, Gallagher L, Thompson L, Khorashad JS, LimbÄck-Stanic C. The KDR (VEGFR-2) Genetic Polymorphism Q472H and c-KIT Polymorphism M541L Are Associated With More Aggressive Behaviour in Astrocytic Gliomas. Cancer Genomics Proteomics 2021; 17:715-727. [PMID: 33099473 DOI: 10.21873/cgp.20226] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND/AIM Better diagnostic and prognostic markers are required for a more accurate diagnosis and an earlier detection of glioma progression and for suggesting better treatment strategies. This retrospective study aimed to identify actionable gene variants to define potential markers of clinical significance. MATERIALS AND METHODS 56 glioblastomas (GBM) and 44 grade 2-3 astrocytomas were profiled with next generation sequencing (NGS) as part of routine diagnostic workup and bioinformatics analysis was used for the identification of variants. CD34 immunohistochemistry (IHC) was used to measure microvessel density (MVD) and Log-rank test to compare survival and progression in the presence or absence of these variants. RESULTS Bioinformatic analysis highlighted frequently occurring variants in genes involved in angiogenesis regulation (KDR, KIT, TP53 and PIK3CA), with the most common ones being KDR (rs1870377) and KIT (rs3822214). The KDR variant was associated with increased MVD and shorter survival in GBM. We did not observe any correlation between the KIT variant and MVD; however, there was an association with tumour grade. CONCLUSION This study highlights the role of single-nucleotide variants (SNVs) that may be considered non-pathogenic and suggests the prognostic significance for survival of KIT rs3822214 and KDR rs1870377 and potential importance in planning new treatment strategies for gliomas.
Collapse
Affiliation(s)
- Niyaz Zaman
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K
| | - Serena Santhana Dass
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K
| | - Persephone DU Parcq
- Department of Cell Pathology, Imperial College Healthcare NHS Trust, London, U.K
| | - Suzanne Macmahon
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Lewis Gallagher
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Lisa Thompson
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, London, U.K
| | - Jamshid S Khorashad
- Department of Immunology and Inflammation, Imperial College London, London, U.K
| | - Clara LimbÄck-Stanic
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, U.K. .,Department of Cell Pathology, Imperial College Healthcare NHS Trust, London, U.K
| |
Collapse
|
8
|
Cruz Da Silva E, Mercier MC, Etienne-Selloum N, Dontenwill M, Choulier L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers (Basel) 2021; 13:1795. [PMID: 33918704 PMCID: PMC8069979 DOI: 10.3390/cancers13081795] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM), the most frequent and aggressive glial tumor, is currently treated as first line by the Stupp protocol, which combines, after surgery, radiotherapy and chemotherapy. For recurrent GBM, in absence of standard treatment or available clinical trials, various protocols including cytotoxic drugs and/or bevacizumab are currently applied. Despite these heavy treatments, the mean overall survival of patients is under 18 months. Many clinical studies are underway. Based on clinicaltrials.org and conducted up to 1 April 2020, this review lists, not only main, but all targeted therapies in phases II-IV of 257 clinical trials on adults with newly diagnosed or recurrent GBMs for the last twenty years. It does not involve targeted immunotherapies and therapies targeting tumor cell metabolism, that are well documented in other reviews. Without surprise, the most frequently reported drugs are those targeting (i) EGFR (40 clinical trials), and more generally tyrosine kinase receptors (85 clinical trials) and (ii) VEGF/VEGFR (75 clinical trials of which 53 involving bevacizumab). But many other targets and drugs are of interest. They are all listed and thoroughly described, on an one-on-one basis, in four sections related to targeting (i) GBM stem cells and stem cell pathways, (ii) the growth autonomy and migration, (iii) the cell cycle and the escape to cell death, (iv) and angiogenesis.
Collapse
Affiliation(s)
- Elisabete Cruz Da Silva
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Marie-Cécile Mercier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Nelly Etienne-Selloum
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
- Service de Pharmacie, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Monique Dontenwill
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Laurence Choulier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| |
Collapse
|
9
|
Chen Y, Guo L, Li X, Liu R, Ren C, Du S. Reduced-dose bevacizumab vs. standard-dose bevacizumab in recurrent high-grade glioma: Which one is better? A meta-analysis. Clin Neurol Neurosurg 2020; 198:106239. [PMID: 33007724 DOI: 10.1016/j.clineuro.2020.106239] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 11/17/2022]
Abstract
BACKGROUND Based on the effective radiological responses, bevacizumab (BEV) has been widely used in the treatment of recurrent high-grade glioma. Although the current standard dose is 5 mg/kg/week, the optimal dosage of BEV is controversial, as few dose-response studies have been performed in recent years. Therefore, we conducted a meta-analysis to explore the value of reduced-dose bevacizumab versus standard-dose bevacizumab in recurrent high-grade glioma treatment. METHODS Three major electronic databases (PubMed, EMBASE and the Cochrane Library) were searched for eligible documents published before February 2020. Literature on low-dose bevacizumab versus conventional dose in progressive high-grade glioma was included, and the endpoints of eligible researches should be progression-free survival (PFS) and overall survival (OS). All available data were collected and then analyzed with Stata software. RESULTS Four cohort studies were evaluated, including 552 patients (reduced-dose BEV group: 257, standard-dose BEV group: 295). Low dose BEV seems to slightly improve survival compared to conventional dose as HR < 1 indicates a protective effect, but no significant differences in OS (HR 0.77; 95 % CI 0.53-1.10; P = 0.151) and PFS (HR 0.66; 95 % CI 0.37-1.20; P = 0.175) were found between the two groups in this study. CONCLUSION Reduced-dose bevacizumab schedule resulted in similar OS and PFS to standard-dose bevacizumab in recurrent high-grade glioma, with less side effects and less cost of treatment. Therefore, low-dose bevacizumab represents a promising therapeutic option for recurrent high-grade glioma patients. Further prospective randomized trials are needed to confirm our results.
Collapse
Affiliation(s)
- Yulei Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Longbin Guo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuanzi Li
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rongping Liu
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Ren
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Shasha Du
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
10
|
Ahir BK, Engelhard HH, Lakka SS. Tumor Development and Angiogenesis in Adult Brain Tumor: Glioblastoma. Mol Neurobiol 2020; 57:2461-2478. [PMID: 32152825 PMCID: PMC7170819 DOI: 10.1007/s12035-020-01892-8] [Citation(s) in RCA: 248] [Impact Index Per Article: 49.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023]
Abstract
Angiogenesis is the growth of new capillaries from the preexisting blood vessels. Glioblastoma (GBM) tumors are highly vascularized tumors, and glioma growth depends on the formation of new blood vessels. Angiogenesis is a complex process involving proliferation, migration, and differentiation of vascular endothelial cells (ECs) under the stimulation of specific signals. It is controlled by the balance between its promoting and inhibiting factors. Various angiogenic factors and genes have been identified that stimulate glioma angiogenesis. Therefore, attention has been directed to anti-angiogenesis therapy in which glioma proliferation is inhibited by inhibiting the formation of new tumor vessels using angiogenesis inhibitory factors and drugs. Here, in this review, we highlight and summarize the various molecular mediators that regulate GBM angiogenesis with focus on recent clinical research on the potential of exploiting angiogenic pathways as a strategy in the treatment of GBM patients.
Collapse
Affiliation(s)
- Bhavesh K Ahir
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Herbert H Engelhard
- Department of Neurosurgery, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA
| | - Sajani S Lakka
- Section of Hematology and Oncology, University of Illinois College of Medicine at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
11
|
Shimizu T, Ishida J, Kurozumi K, Ichikawa T, Otani Y, Oka T, Tomita Y, Hattori Y, Uneda A, Matsumoto Y, Date I. δ-Catenin Promotes Bevacizumab-Induced Glioma Invasion. Mol Cancer Ther 2019; 18:812-822. [PMID: 30872378 DOI: 10.1158/1535-7163.mct-18-0138] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 09/26/2018] [Accepted: 02/22/2019] [Indexed: 11/16/2022]
Abstract
The combination of bevacizumab with temozolomide and radiotherapy was shown to prolong progression-free survival in newly diagnosed patients with glioblastoma, and this emphasizes the potential of bevacizumab as a glioma treatment. However, although bevacizumab effectively inhibits angiogenesis, it has also been reported to induce invasive proliferation. This study examined gene expression in glioma cells to investigate the mechanisms of bevacizumab-induced invasion. We made a human glioma U87ΔEGFR cell xenograft model by stereotactically injecting these cells into the brain of animals. We administered bevacizumab intraperitoneally three times per week. At 18 days after tumor implantation, the brains were removed for histopathology and mRNA was extracted. In vivo, bevacizumab treatment increased glioma cell invasion. qRT-PCR array analysis revealed upregulation of δ-catenin (CTNND2) and several other factors. In vitro, bevacizumab treatment upregulated δ-catenin expression. A low concentration of bevacizumab was not cytotoxic, but tumor cell motility was increased in scratch wound assays and two-chamber assays. Overexpression of δ-catenin increased the tumor invasion in vitro and in vivo However, δ-catenin knockdown decreased glioma cell invasiveness. The depth of tumor invasion in the U87ΔEGFR cells expressing δ-catenin was significantly increased compared with empty vector-transfected cells. The increase in invasive capacity induced by bevacizumab therapy was associated with upregulation of δ-catenin expression in invasive tumor cells. This finding suggests that δ-catenin is related to tumor invasion and migration.
Collapse
Affiliation(s)
- Toshihiko Shimizu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Joji Ishida
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tetsuo Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuhiko Hattori
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuhito Uneda
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuji Matsumoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
12
|
Cayrol F, Sterle HA, Díaz Flaqué MC, Barreiro Arcos ML, Cremaschi GA. Non-genomic Actions of Thyroid Hormones Regulate the Growth and Angiogenesis of T Cell Lymphomas. Front Endocrinol (Lausanne) 2019; 10:63. [PMID: 30814977 PMCID: PMC6381017 DOI: 10.3389/fendo.2019.00063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/23/2019] [Indexed: 12/16/2022] Open
Abstract
T-cell lymphomas (TCL) are a heterogeneous group of aggressive clinical lymphoproliferative disorders with considerable clinical, morphological, immunophenotypic, and genetic variation, including ~10-15% of all lymphoid neoplasms. Several evidences indicate an important role of the non-neoplastic microenvironment in promoting both tumor growth and dissemination in T cell malignancies. Thus, dysregulation of integrin expression and activity is associated with TCL survival and proliferation. We found that thyroid hormones acting via the integrin αvβ3 receptor are crucial factors in tumor microenvironment (TME) affecting the pathophysiology of TCL cells. Specifically, TH-activated αvβ3 integrin signaling promoted TCL proliferation and induced and an angiogenic program via the up-regulation of the vascular endothelial growth factor (VEGF). This was observed both on different TCL cell lines representing the different subtypes of human hematological malignancy, and in preclinical models of TCL tumors xenotransplanted in immunodeficient mice as well. Moreover, development of solid tumors by inoculation of murine TCLs in syngeneic hyperthyroid mice, showed increased tumor growth along with increased expression of cell cycle regulators. The genomic or pharmacological inhibition of integrin αvβ3 decreased VEGF production, induced TCL cell death and decreased in vivo tumor growth and angiogenesis. Here, we review the non-genomic actions of THs on TCL regulation and their contribution to TCL development and evolution. These actions not only provide novel new insights on the endocrine modulation of TCL, but also provide a potential molecular target for its treatment.
Collapse
Affiliation(s)
- Florencia Cayrol
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Helena A Sterle
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Maria Celeste Díaz Flaqué
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Maria Laura Barreiro Arcos
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Graciela A Cremaschi
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Ciencias Médicas, Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
- Laboratorio de Radioisótopos, Cátedra de Física, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
13
|
Sulforaphane from Cruciferous Vegetables: Recent Advances to Improve Glioblastoma Treatment. Nutrients 2018; 10:nu10111755. [PMID: 30441761 PMCID: PMC6267435 DOI: 10.3390/nu10111755] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 02/07/2023] Open
Abstract
Sulforaphane (SFN), an isothiocyanate (ITC) derived from cruciferous vegetables, particularly broccoli and broccoli sprouts, has been widely investigated due to its promising health-promoting properties in disease, and low toxicity in normal tissue. Although not yet fully understood, many mechanisms of anticancer activity at each step of cancer development have been attributed to this ITC. Given the promising data available regarding SFN, this review aimed to provide an overview on the potential activities of SFN related to the cellular mechanisms involved in glioblastoma (GBM) progression. GBM is the most frequent malignant brain tumor among adults and is currently an incurable disease due mostly to its highly invasive phenotype, and the poor efficacy of the available therapies. Despite all efforts, the median overall survival of GBM patients remains approximately 1.5 years under therapy. Therefore, there is an urgent need to provide support for translating the progress in understanding the molecular background of GBM into more complex, but promising therapeutic strategies, in which SFN may find a leading role.
Collapse
|
14
|
Balça-Silva J, Matias D, Carmo AD, Sarmento-Ribeiro AB, Lopes MC, Moura-Neto V. Cellular and molecular mechanisms of glioblastoma malignancy: Implications in resistance and therapeutic strategies. Semin Cancer Biol 2018; 58:130-141. [PMID: 30266571 DOI: 10.1016/j.semcancer.2018.09.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/12/2018] [Accepted: 09/20/2018] [Indexed: 02/01/2023]
Abstract
Glioblastoma (GB) is the more frequent and malignant brain tumour. In spite of all efforts, the median overall survival of GB patients remains approximately 15 months under therapy. The molecular biology underlying GB is complex, which highlight the need of specific treatment strategies. In fact, the deregulation of several molecular signalling pathways, the existence of the blood-brain barrier (BBB), that makes almost all the chemotherapeutic agents inaccessible to the tumour site, and the existence of a population of stem-like cells known to be responsible for tumour recurrence after therapy, can contribute to GB chemoresistance. In the present review, we summarize the reliable factors responsible for the failure of the most important chemotherapeutic agents in GB. Specifically, we describe the utmost important characteristics of the BBB, as well as the genetic, molecular and transcription factors alterations that lead to tumour malignancy, and ultimately their impact on stem-like cell plasticity modulation. Recently, nanocarriers have attracted increasing attention in brain- and tumour-targeted drug-delivery systems, owing to their potential ability to target cell surface specific molecules and to cross the BBB delivering the drug specifically to the tumour cells, improving efficacy and thus reducing non-specific toxicity. In this sense, we will lastly highlight the therapeutic challenges and improvements regarding GB treatment.
Collapse
Affiliation(s)
- Joana Balça-Silva
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal; Faculty of Medicine, University of Coimbra (FMUC), Coimbra, Portugal; Instituto Estadual do Cérebro Paulo Niemeyer (IECPN) - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.
| | - Diana Matias
- Instituto Estadual do Cérebro Paulo Niemeyer (IECPN) - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil; Instituto de Ciências Biomédicas da Universidade Federal do Rio de Janeiro (ICB-UFRJ), Rio de Janeiro, Brazil.
| | - Anália do Carmo
- Clinical Pathology Department, Coimbra Hospital and Universitary Center (CHUC), Coimbra, Portugal; Center for Neuroscience and Cell Biology, Institute for Biomedical Imaging and Life Sciences (CNC.IBILI) Coimbra, Portugal.
| | - Ana Bela Sarmento-Ribeiro
- Faculty of Medicine, University of Coimbra (FMUC) and Coimbra Institute for Clinical and Biomedical Research (iCBR), group of Environment, Genetics and Oncobiology (CIMAGO), Coimbra, Portugal; Centro Hospitalar Universitário de Coimbra (CHUC), Coimbra, Portugal; Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.
| | - Maria Celeste Lopes
- Center for Neuroscience and Cell Biology and Institute for Biomedical Imaging and Life Sciences (CNC.IBILI), Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra (FFUC); Coimbra, Portugal.
| | - Vivaldo Moura-Neto
- Instituto Estadual do Cérebro Paulo Niemeyer (IECPN) - Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.
| |
Collapse
|
15
|
Mikkelsen VE, Stensjøen AL, Granli US, Berntsen EM, Salvesen Ø, Solheim O, Torp SH. Angiogenesis and radiological tumor growth in patients with glioblastoma. BMC Cancer 2018; 18:862. [PMID: 30176826 PMCID: PMC6122710 DOI: 10.1186/s12885-018-4768-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The preoperative growth of human glioblastomas (GBMs) has been shown to vary among patients. In animal studies, angiogenesis has been linked to hypoxia and faster growth of GBM, however, its relation to the growth of human GBMs is sparsely studied. We have therefore aimed to look for associations between radiological speed of growth and microvessel density (MVD) counts of the endothelial markers vWF (Factor VIII related antigen) and CD105 (endoglin). METHODS Preoperative growth was estimated from segmented tumor volumes of two preoperative T1-weighted postcontrast magnetic resonance imaging scans taken ≥14 days apart in patients with newly diagnosed GBMs. A Gompertzian growth curve was computed from the volume data and separated the patients into two groups of either faster or slower tumor growth than expected. MVD counts of the immunohistochemical markers von Willebrand factor (vWF) (a pan-endothelial marker) and CD105 (a marker of proliferating endothelial cells) were assessed for associations with fast-growing tumors using Mann-Whitney U tests and a multivariable binary logistic regression analysis. RESULTS We found that only CD105-MVD was significantly associated with faster growth in a univariable analysis (p = 0.049). However, CD105-MVD was no longer significant when corrected for the presence of thromboses and high cellular density in a multivariable model, where the latter features were significant independent predictors of faster growth with respective odds ratios 4.2 (95% confidence interval, 1.2, 14.3), p = 0.021 and 2.6 (95% confidence interval, 1.0, 6.5), p = 0.048. CONCLUSIONS MVDs of neither endothelial marker were independently associated with faster growth, suggesting angiogenesis-independent processes contribute to faster glioblastoma growth.
Collapse
Affiliation(s)
- Vilde Elisabeth Mikkelsen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.
| | - Anne Line Stensjøen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway
| | - Unn Sophie Granli
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.,Cellular and Molecular Imaging Core Facility (CMIC), Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erik Magnus Berntsen
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.,Department of Radiology and Nuclear Medicine, St. Olavs University Hospital, Trondheim, Norway
| | - Øyvind Salvesen
- Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Ole Solheim
- Department of Neurosurgery, St. Olavs University Hospital, Trondheim, Norway.,National Advisory Unit for Ultrasound and Image Guided Therapy, St. Olavs University Hospital, Trondheim, Norway.,Department of Neuromedicine and Movement Science, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre Helge Torp
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Erling Skjalgssons gate 1, 7030, Trondheim, Norway.,Department of Pathology, St. Olavs University Hospital, Trondheim, Norway
| |
Collapse
|
16
|
Oka T, Kurozumi K, Shimazu Y, Ichikawa T, Ishida J, Otani Y, Shimizu T, Tomita Y, Sakaguchi M, Watanabe M, Nasu Y, Kumon H, Date I. A super gene expression system enhances the anti-glioma effects of adenovirus-mediated REIC/Dkk-3 gene therapy. Sci Rep 2016; 6:33319. [PMID: 27625116 PMCID: PMC5022040 DOI: 10.1038/srep33319] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/24/2016] [Indexed: 12/26/2022] Open
Abstract
Reduced expression in immortalized cells/Dickkopf-3 (REIC/Dkk-3) is a tumor suppressor and therapeutic gene in many human cancers. Recently, an adenovirus REIC vector with the super gene expression system (Ad-SGE-REIC) was developed to increase REIC/Dkk-3 expression and enhance therapeutic effects compared with the conventional adenoviral vector (Ad-CAG-REIC). In this study, we investigated the in vitro and in vivo effects of Ad-SGE-REIC on malignant glioma. In U87ΔEGFR and GL261 glioma cells, western blotting confirmed that robust upregulation of REIC/Dkk-3 expression occurred in Ad-SGE-REIC-transduced cells, most notably after transduction at a multiplicity of infection of 10. Cytotoxicity assays showed that Ad-SGE-REIC resulted in a time-dependent and significant reduction in the number of malignant glioma cells attaching to the bottom of culture wells. Xenograft and syngeneic mouse intracranial glioma models treated with Ad-SGE-REIC had significantly longer survival than those treated with the control vector Ad-LacZ or with Ad-CAG-REIC. This study demonstrated the anti-glioma effect of Ad-SGE-REIC, which may represent a promising strategy for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Tetsuo Oka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yosuke Shimazu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Joji Ishida
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Toshihiko Shimizu
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yusuke Tomita
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Masami Watanabe
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromi Kumon
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
17
|
Adhesion molecules and the extracellular matrix as drug targets for glioma. Brain Tumor Pathol 2016; 33:97-106. [PMID: 26992378 DOI: 10.1007/s10014-016-0261-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/07/2016] [Indexed: 12/14/2022]
Abstract
The formation of tumor vasculature and cell invasion along white matter tracts have pivotal roles in the development and progression of glioma. A better understanding of the mechanisms of angiogenesis and invasion in glioma will aid the development of novel therapeutic strategies. The processes of angiogenesis and invasion cause the production of an array of adhesion molecules and extracellular matrix (ECM) components. This review focuses on the role of adhesion molecules and the ECM in malignant glioma. The results of clinical trials using drugs targeted against adhesion molecules and the ECM for glioma are also discussed.
Collapse
|
18
|
Zhang Z, Li C, Shang L, Zhang Y, Zou R, Zhan Y, Bi B. Sulforaphane induces apoptosis and inhibits invasion in U251MG glioblastoma cells. SPRINGERPLUS 2016; 5:235. [PMID: 27026929 PMCID: PMC4771656 DOI: 10.1186/s40064-016-1910-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 02/17/2016] [Indexed: 12/18/2022]
Abstract
In recent studies, sulforaphane (SFN) has been seen to demonstrate antioxidant and anti-tumor activities. In the present study, the viability inhibition effects of SFN in U251MG glioblastoma cells were analyzed by MTS. Morphology changes were observed by microscope. Apoptotic effects of SFN were evaluated by annexin V binding capacity with flow cytometric analysis. Invasion inhibition effects of SFN were tested by the invasion assay. The molecular mechanisms of apoptotic effects and invasion inhibition effects of SFN were detected by western blot and gelatin zymography. The results indicated that SFN has potent apoptotic effects and invasion inhibition effects against U251MG glioblastoma cells. These effects are both dose dependent. Taken together, SFN possessed apoptotic activity on U251MG cells indicated by increased annexin V-binding capacity, Bad, Bax, cytochrome C expression, and decreased Bcl-2 and survivin expressions. SFN inhibited invasion in U251MG cells via upregulation of E-cadherin and downregulation of MMP-2, MMP-9 and Galectin-3.
Collapse
Affiliation(s)
- Zhen Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Chunliu Li
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Li Shang
- Affiliated Hospital of Binzhou Medical University, Binzhou, China
| | | | - Rong Zou
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | - Yan Zhan
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100 China
| | | |
Collapse
|
19
|
Popescu AM, Purcaru SO, Alexandru O, Dricu A. New perspectives in glioblastoma antiangiogenic therapy. Contemp Oncol (Pozn) 2015; 20:109-18. [PMID: 27358588 PMCID: PMC4925727 DOI: 10.5114/wo.2015.56122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/15/2015] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GB) is highly vascularised tumour, known to exhibit enhanced infiltrative potential. One of the characteristics of glioblastoma is microvascular proliferation surrounding necrotic areas, as a response to a hypoxic environment, which in turn increases the expression of angiogenic factors and their signalling pathways (RAS/RAF/ERK/MAPK pathway, PI3K/Akt signalling pathway and WTN signalling cascade). Currently, a small number of anti-angiogenic drugs, extending glioblastoma patients survival, are available for clinical use. Most medications are ineffective in clinical therapy of glioblastoma due to acquired malignant cells or intrinsic resistance, angiogenic receptors cross-activation and redundant intracellular signalling, or the inability of the drug to cross the blood-brain barrier and to reach its target in vivo. Researchers have also observed that GB tumours are different in many aspects, even when they derive from the same tissue, which is the reason for personalised therapy. An understanding of the molecular mechanisms regulating glioblastoma angiogenesis and invasion may be important in the future development of curative therapeutic approaches for the treatment of this devastating disease.
Collapse
Affiliation(s)
| | - Stefana Oana Purcaru
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova and Clinical Hospital of Neuropsychiatry Craiova, Craiova, Romania
| | - Anica Dricu
- Unit of Biochemistry, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| |
Collapse
|
20
|
Recent advance in molecular angiogenesis in glioblastoma: the challenge and hope for anti-angiogenic therapy. Brain Tumor Pathol 2015; 32:229-36. [PMID: 26437643 DOI: 10.1007/s10014-015-0233-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 09/23/2015] [Indexed: 12/18/2022]
Abstract
Glioblastoma (GBM) is the most highly malignant brain tumor in the human central nerve system. In this paper, we review new and significant molecular findings on angiogenesis and possible resistance mechanisms. Expression of a number of genes and regulators has been shown to be upregulated in GBM microvessel cells, such as interleukin-8, signal transducer and activator of transcription 3, Tax-interacting protein-1, hypoxia induced factor-1 and anterior gradient protein 2. The regulator factors that may strongly promote angiogenesis by promoting endothelial cell metastasis, changing the microenvironment, enhancing the ability of resistance to anti-angiogenic therapy, and that inhibit angiogenesis are reviewed. Based on the current knowledge, several potential targets and strategies are proposed for better therapeutic outcomes, such as its mRNA interference of DII4-Notch signaling pathway and depletion of b1 integrin expression. We also discuss possible mechanisms underlying the resistance to anti-angiogenesis and future directions and challenges in developing new targeted therapy for GBM.
Collapse
|
21
|
Fakhoury M. Drug delivery approaches for the treatment of glioblastoma multiforme. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2015; 44:1365-73. [PMID: 26046399 DOI: 10.3109/21691401.2015.1052467] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CONTEXT Glioblastoma multiforme (GBM) is by far the most common and aggressive form of glial tumor. It is characterized by a highly proliferative population of cells that invade surrounding tissue and that frequently recur after surgical resection and chemotherapy. Over the last decades, a number of promising novel pharmacological approaches have been investigated, but most of them have failed clinical trials due to some side-effects such as toxicity and poor drug delivery to the brain. The major obstacle in the treatment of GBM is the presence of the blood-brain barrier (BBB). Due to their relatively high molecular weight, most therapeutic drugs fail to cross the BBB from the blood circulation. OBJECTIVE This paper sheds light on the characteristics of GBM and the challenges of current pharmacological treatments. A closer look is given to the role of nanotechnology in the field of drug delivery, and its application in the treatment of brain tumors such as GBM. METHOD For this purpose, effort was made to select the most recent studies using predefined search criteria that included at least one of the following keywords in the PubMed and Medline databases: glioblastoma, drug delivery, blood-brain barrier, nanotechnology, and nanoparticle. CONCLUSION Breakthrough in nanotechnology offers promising applications in cancer therapy and targeted drug delivery. However, more efforts need to be devoted to the development of novel therapeutic strategies that enable the delivery of drugs to desired areas of the brain with limited side-effects and higher therapeutic efficiency.
Collapse
Affiliation(s)
- Marc Fakhoury
- a Department of Neurosciences , University of Montreal , Montreal , QC , Canada
| |
Collapse
|
22
|
Alifieris C, Trafalis DT. Glioblastoma multiforme: Pathogenesis and treatment. Pharmacol Ther 2015; 152:63-82. [PMID: 25944528 DOI: 10.1016/j.pharmthera.2015.05.005] [Citation(s) in RCA: 528] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 04/28/2015] [Indexed: 12/12/2022]
Abstract
Each year, about 5-6 cases out of 100,000 people are diagnosed with primary malignant brain tumors, of which about 80% are malignant gliomas (MGs). Glioblastoma multiforme (GBM) accounts for more than half of MG cases. They are associated with high morbidity and mortality. Despite current multimodality treatment efforts including maximal surgical resection if feasible, followed by a combination of radiotherapy and/or chemotherapy, the median survival is short: only about 15months. A deeper understanding of the pathogenesis of these tumors has presented opportunities for newer therapies to evolve and an expectation of better control of this disease. Lately, efforts have been made to investigate tumor resistance, which results from complex alternate signaling pathways, the existence of glioma stem-cells, the influence of the blood-brain barrier as well as the expression of 0(6)-methylguanine-DNA methyltransferase. In this paper, we review up-to-date information on MGs treatment including current approaches, novel drug-delivering strategies, molecular targeted agents and immunomodulative treatments, and discuss future treatment perspectives.
Collapse
Affiliation(s)
| | - Dimitrios T Trafalis
- Laboratory of Pharmacology, Medical School, University of Athens, Athens, Greece.
| |
Collapse
|
23
|
Integrin inhibitor suppresses bevacizumab-induced glioma invasion. Transl Oncol 2014; 7:292-302.e1. [PMID: 24704537 PMCID: PMC4101347 DOI: 10.1016/j.tranon.2014.02.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 02/03/2014] [Accepted: 02/10/2014] [Indexed: 12/30/2022] Open
Abstract
Glioblastoma is known to secrete high levels of vascular endothelial growth factor (VEGF), and clinical studies with bevacizumab, a monoclonal antibody to VEGF, have demonstrated convincing therapeutic benefits in glioblastoma patients. However, its induction of invasive proliferation has also been reported. We examined the effects of treatment with cilengitide, an integrin inhibitor, on bevacizumab-induced invasive changes in glioma. U87ΔEGFR cells were stereotactically injected into the brain of nude mice or rats. Five days after tumor implantation, cilengitide and bevacizumab were administered intraperitoneally three times a week. At 18 days after tumor implantation, the brains were removed and observed histopathologically. Next, the bevacizumab and cilengitide combination group was compared to the bevacizumab monotherapy group using microarray analysis. Bevacizumab treatment led to increased cell invasion in spite of decreased angiogenesis. When the rats were treated with a combination of bevacizumab and cilengitide, the depth of tumor invasion was significantly less than with only bevacizumab. Pathway analysis demonstrated the inhibition of invasion-associated genes such as the integrin-mediated cell adhesion pathway in the combination group. This study showed that the combination of bevacizumab with cilengitide exerted its anti-invasive effect. The elucidation of this mechanism might contribute to the treatment of bevacizumab-refractory glioma.
Collapse
|