1.Top 100 most cited articles on awake glioma surgery: A bibliometric analysis.
Mary Angeline Luz U. HERNANDEZ ; Almira Doreen Abigail O. APOR ; Kevin Ivan P. CHAN ; Kathleen Joy O. KHU
Acta Medica Philippina 2025;59(13):60-73
BACKGROUND AND OBJECTIVE
Awake craniotomy is often used in the surgery of glioma, the most common primary brain tumor. It has been proven to maximize the extent of tumor resection while minimizing post-operative neurologic deficits. Extensive research has been conducted on this topic, and we would like to perform a bibliometric analysis to identify the top 100 most cited articles in awake glioma surgery. Knowing the relevant and most impactful studies in the field would help clinicians streamline the evidence and determine its application in their practice.
METHODSIn October 2023, we performed a title-specific search on the Scopus and PubMed databases using (“glioma*” OR “astrocytoma*” OR “glioblastoma” OR “low grade glioma” OR “high grade glioma”) and (“awake craniotomy” OR “awake surgery” OR “awake brain surgery” OR “awake neurosurgery”) as our query term without any restriction criteria. The top 100 most cited articles were identified, reviewed, and analyzed.
RESULTSOur search yielded a total of 5557 articles published. The top article had a citation count of 834 and reported on functional outcome after language mapping in glioma resection. Journal of Neurosurgery had the most number of publications. Neurosurgeons (n=81) were the primary author in most publications, followed by anesthesiologists (n=22) and neurologists (n=6). Three countries (USA, France, Italy) contributed to 74% of the articles. Most of the articles were reviews and case reports/series.
CONCLUSIONThis study identified the top 100 most cited articles on awake glioma surgery. The content dealt with several aspects of awake craniotomy such as brain mapping, intraoperative techniques and adjuncts, and practice recommendations. This analysis can help identify knowledge gaps and potential areas of research in glioma surgery.
Glioma ; Glioblastoma ; Astrocytoma ; Bibliometric Analysis ; Bibliometrics
2.Nodular melanoma in a 53-year-old male with glioblastoma multiforme: A rare case report
Arbie Sofia P. Merilleno ; Mary Elizabeth S. Danga ; Alma Gay Concepcion T. Amado
Acta Medica Philippina 2024;58(3):82-86
Although melanoma only accounts for 1% of skin cancers, it is responsible for most skin cancer deaths. Glioblastoma multiforme, a high-grade astrocytoma, is the most aggressive and devastating primary brain tumor. These two diseases remain to be the biggest therapeutic challenge in both specialties of dermatology and neuro-oncology.
A 53-year-old Filipino male who presented with a 2-year history of generalized dark brown and black patches on the body developed weakness and numbness of the left extremities. Biopsy and immunohistochemical staining of the skin revealed nodular melanoma with adjacent regressing melanoma. Biopsy of the intracranial mass showed glioblastoma multiforme. One month after the partial excision of the intracranial mass, the patient expired due to brain herniation.
Nodular melanoma and glioblastoma multiforme may occur concomitantly in a patient. A review of the literature
suggests a shared genetic predisposition. Its existence carries a poor prognosis and requires early detection to start aggressive treatment.
Melanoma
;
Glioma
;
Glioblastoma
;
Association
3.Recurrent epithelioid glioblastoma in a young patient with systemic lupus erythematosus: A case report
Renato C. Galvan, Jr. ; Paul Vincent A. Opinaldo ; Ma. Luisa Gwenn F. Pabellano-Tiongson
Philippine Journal of Neurology 2024;27(1):15-19
Glioblastoma multiforme (GBM)
represents the most malignant form of brain
tumor and is relatively common, comprising
nearly almost 20% of all primary malignancies of the central nervous system1.
GBM is a WHO grade IV tumor with several
variants, depending primarily on their genetic
signature and on the predominant histological
architecture. Among the variants of GBM,
epithelioid glioblastoma (E-GBM) has been
one of the more recently described. This
tumor, documented to be highly malignant
and clinically aggressive, has been separated
from close variants and thus differentials,
pleomorphic anaplastic xanthoastrocytoma,
rhabdoid GBM, small cell and giant cell GBM,
GBM with neuroectodermal differentiation,
and gliosarcoma2.
Autoimmune diseases have been
linked within creased risk of CNS
complications, from the constant effects of
chronic inflammatory milieu. Systemic lupus
erythematosus (SLE) has been associated with
several CNS abnormalities, hence the terms
CNS lupus or neuropsychiatric lupus.
Likewise, SLE has been repeatedly associated
with CNS malignancies in several cases and
case reports.
To date, there is paucity in the
reported cases of malignant brain tumors,
especially rare variants, in patients with SLE.
While it is hypothesized that the
inflammatory milieu that bathes the brain in a
dynamic microenvironment that influences
the incidence of rare variants of GBM, clinicians should be mindful, as treatment is
challenging: it may either induce exacerbation
of autoimmunity or cause undertreatment of
the malignancy. This complex interplay births
curiosity into the enigma of autoimmunity
and oncology.
In this particular report, we highlight the case
of a patient with SLE who developed E-GBM.
We identify the clinicopathologic features of
the tumor present in the patient and explore
the known aspects of the crosstalk between
SLE and E-GBM.
Lupus Erythematosus, Systemic
;
Glioblastoma
4.A case of Constitutional Mismatch Repair Deficiency (CMMRD)
Ma. Janina T. Layosa ; Julieta Rita Gonzales-Santos
Journal of the Philippine Medical Association 2023;102(1):75-81
Constitutionalmismatch repair deficiency(CMMRD) is a hereditary predisposition of malignancy evident in childhood leukemias, lymphomas, and malignant tumors of the brain, GI tract. It is a very rare condition that affects 1 per 1 million patients. Patients with CMMRD syndrome may also manifest with Neurofibromatosis Type 1 (NF1) phenotypic features, and benign masses, particularly in the gastrointestinal tract. This is a case of a 12-year old male who presented with phenotypic features of NF1, developed Acute Lymphoblastic Leukemia at 7 years old and went into remission. He subsequently developed synchronous Glioblastoma and Poorly differentiated Adenocarcinoma of the rectum.This report aims to raise awareness regarding the possibility of a CMMRD syndrome in pediatric patients who present with phenotypic features of NF1, and in those patients who present with two or more malignancies in their lifetime.
Glioblastoma
;
Precursor Cell Lymphoblastic Leukemia-Lymphoma
5.Construction and functional analysis of EGFRvIII CAR-T cells co-expressing IL-15 and CCL19.
Wanqiong CHEN ; Na XIAN ; Shaomei LIN ; Wanting LIAO ; Mingzhu CHEN
Chinese Journal of Biotechnology 2023;39(9):3787-3799
The aim of this study was to investigate the functional characteristics and in vitro specific killing effect of EGFRvIII CAR-T cells co-expressing interleukin-15 and chemokine CCL19, in order to optimize the multiple functions of CAR-T cells and improve the therapeutic effect of CAR-T cells targeting EGFRvIII on glioblastoma (GBM). The recombinant lentivirus plasmid was obtained by genetic engineering, transfected into 293T cells to obtain lentivirus and infected T cells to obtain the fourth generation CAR-T cells targeting EGFRvIII (EGFRvIII-IL-15-CCL19 CAR-T). The expression rate of CAR molecules, proliferation, chemotactic ability, in vitro specific killing ability and anti-apoptotic ability of the fourth and second generation CAR-T cells (EGFRvIII CAR-T) were detected by flow cytometry, cell counter, chemotaxis chamber and apoptosis kit. The results showed that compared with EGFRvIII CAR-T cells, EGFRvIII-IL-15-CCL19 CAR-T cells successfully secreted IL-15 and CCL19, and had stronger proliferation, chemotactic ability and anti-apoptosis ability in vitro (all P < 0.05), while there was no significant difference in killing ability in vitro. Therefore, CAR-T cells targeting EGFRvIII and secreting IL-15 and CCL19 are expected to improve the therapeutic effect of glioblastoma and provide an experimental basis for clinical trials.
Humans
;
Receptors, Chimeric Antigen/metabolism*
;
Glioblastoma/metabolism*
;
Interleukin-15/metabolism*
;
Chemokine CCL19/metabolism*
;
Cell Line, Tumor
;
T-Lymphocytes/metabolism*
6.FOXO1-miR-506 axis promotes chemosensitivity to temozolomide and suppresses invasiveness in glioblastoma through a feedback loop of FOXO1/miR-506/ETS1/FOXO1.
Chao CHEN ; Yu'e LIU ; Hongxiang WANG ; Xu ZHANG ; Yufeng SHI ; Juxiang CHEN
Journal of Zhejiang University. Science. B 2023;24(8):698-710
To explore the role of forkhead box protein O1 (FOXO1) in the progression of glioblastoma multiforme (GBM) and related drug resistance, we deciphered the roles of FOXO1 and miR-506 in proliferation, apoptosis, migration, invasion, autophagy, and temozolomide (TMZ) sensitivity in the U251 cell line using in vitro and in vivo experiments. Cell viability was tested by a cell counting kit-8 (CCK8) kit; migration and invasion were checked by the scratching assay; apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining and flow cytometry. The construction of plasmids and dual-luciferase reporter experiment were carried out to find the interaction site between FOXO1 and miR-506. Immunohistochemistry was done to check the protein level in tumors after the in vivo experiment. We found that the FOXO1-miR-506 axis suppresses GBM cell invasion and migration and promotes GBM chemosensitivity to TMZ, which was mediated by autophagy. FOXO1 upregulates miR-506 by binding to its promoter to enhance transcriptional activation. MiR-506 could downregulate E26 transformation-specific 1 (ETS1) expression by targeting its 3'-untranslated region (UTR). Interestingly, ETS1 promoted FOXO1 translocation from the nucleus to the cytosol and further suppressed the FOXO1-miR-506 axis in GBM cells. Consistently, both miR-506 inhibition and ETS1 overexpression could rescue FOXO1 overactivation-mediated TMZ chemosensitivity in mouse models. Our study demonstrated a negative feedback loop of FOXO1/miR-506/ETS1/FOXO1 in GBM in regulating invasiveness and chemosensitivity. Thus, the above axis might be a promising therapeutic target for GBM.
Animals
;
Mice
;
Brain Neoplasms/genetics*
;
Cell Line, Tumor
;
Cell Proliferation
;
Drug Resistance, Neoplasm
;
Feedback
;
Gene Expression Regulation, Neoplastic
;
Glioblastoma/metabolism*
;
MicroRNAs/metabolism*
;
Temozolomide/therapeutic use*
;
Humans
;
Forkhead Box Protein O1/metabolism*
7.Leucine-rich repeats containing 4 protein (LRRC4) in memory, psychoneurosis, and glioblastoma.
Chinese Medical Journal 2023;136(1):4-12
Leucine-rich repeats containing 4 ( LRRC4 , also named netrin-G ligand 2 [NGL-2]) is a member of the NetrinGs ligands (NGLs) family. As a gene with relatively high and specific expression in brain, it is a member of the leucine-rich repeat superfamily and has been proven to be a suppressor gene for gliomas, thus being involved in gliomagenesis. LRRC4 is the core of microRNA-dependent multi-phase regulatory loops that inhibit the proliferation and invasion of glioblastoma (GB) cells, including LRRC4/NGL2-activator protein 2 (AP2)-microRNA (miR) 182-LRRC4 and LRRC4-miR185-DNA methyltransferase 1 (DNMT1)-LRRC4/specific protein 1 (SP1)-DNMT1-LRRC4. In this review, we demonstrated LRRC4 as a new member of the partitioning-defective protein (PAR) polarity complex that promotes axon differentiation, mediates the formation and plasticity of synapses, and assists information input to the hippocampus and storage of memory. As an important synapse regulator, aberrant expression of LRRC4 has been detected in autism, spinal injury and GBs. LRRC4 is a candidate susceptibility gene for autism and a neuro-protective factor in spinal nerve damage. In GBs, LRRC4 is a novel inhibitor of autophagy, and an inhibitor of protein-protein interactions involving in temozolomide resistance, tumor immune microenvironment, and formation of circular RNA.
Humans
;
Cell Line, Tumor
;
Glioblastoma/metabolism*
;
Leucine
;
Leucine-Rich Repeat Proteins/genetics*
;
MicroRNAs
;
Nerve Tissue Proteins/genetics*
;
Tumor Microenvironment
8.Effect of Morus alba extract sanggenon C on growth and proliferation of glioblastoma cells.
Wen-Han TANG ; Zhi-Ning ZHANG ; Hua-Rui CAI ; Wei SUN ; He YANG ; Er-Hu ZHAO ; Hong-Juan CUI
China Journal of Chinese Materia Medica 2023;48(1):211-219
Glioblastoma is the most common primary cranial malignancy, and chemotherapy remains an important tool for its treatment. Sanggenon C(San C), a class of natural flavonoids extracted from Morus plants, is a potential antitumor herbal monomer. In this study, the effect of San C on the growth and proliferation of glioblastoma cells was examined by methyl thiazolyl tetrazolium(MTT) assay and 5-bromodeoxyuridinc(BrdU) labeling assay. The effect of San C on the tumor cell cycle was examined by flow cytometry, and the effect of San C on clone formation and self-renewal ability of tumor cells was examined by soft agar assay. Western blot and bioinformatics analysis were used to investigate the mechanism of the antitumor activity of San C. In the presence of San C, the MTT assay showed that San C significantly inhibited the growth and proliferation of tumor cells in a dose and time-dependent manner. BrdU labeling assay showed that San C significantly attenuated the DNA replication activity in the nucleus of tumor cells. Flow cytometry confirmed that San C blocked the cell cycle of tumor cells in G_0/G_1 phase. The soft agar clone formation assay revealed that San C significantly attenuated the clone formation and self-renewal ability of tumor cells. The gene set enrichment analysis(GSEA) implied that San C inhibited the tumor cell division cycle by affecting the myelocytomatosis viral oncogene(MYC) signaling pathway. Western blot assay revealed that San C inhibited the expression of cyclin through the regulation of the MYC signaling pathway by lysine demethylase 4B(KDM4B), which ultimately inhibited the growth and proliferation of glioblastoma cells and self-renewal. In conclusion, San C exhibits the potential antitumor activity by targeting the KDM4B-MYC axis to inhibit glioblastoma cell growth, proliferation, and self-renewal.
Humans
;
Glioblastoma/genetics*
;
Bromodeoxyuridine/therapeutic use*
;
Signal Transduction
;
Proto-Oncogene Proteins c-myc/metabolism*
;
Agar
;
Cell Proliferation
;
Cell Line, Tumor
;
Apoptosis
;
Jumonji Domain-Containing Histone Demethylases/metabolism*
9.Hypoxia-induced ROS aggravate tumor progression through HIF-1α-SERPINE1 signaling in glioblastoma.
Lin ZHANG ; Yuanyuan CAO ; Xiaoxiao GUO ; Xiaoyu WANG ; Xiao HAN ; Kouminin KANWORE ; Xiaoliang HONG ; Han ZHOU ; Dianshuai GAO
Journal of Zhejiang University. Science. B 2023;24(1):32-49
Hypoxia, as an important hallmark of the tumor microenvironment, is a major cause of oxidative stress and plays a central role in various malignant tumors, including glioblastoma. Elevated reactive oxygen species (ROS) in a hypoxic microenvironment promote glioblastoma progression; however, the underlying mechanism has not been clarified. Herein, we found that hypoxia promoted ROS production, and the proliferation, migration, and invasion of glioblastoma cells, while this promotion was restrained by ROS scavengers N-acetyl-L-cysteine (NAC) and diphenyleneiodonium chloride (DPI). Hypoxia-induced ROS activated hypoxia-inducible factor-1α (HIF-1α) signaling, which enhanced cell migration and invasion by epithelial-mesenchymal transition (EMT). Furthermore, the induction of serine protease inhibitor family E member 1 (SERPINE1) was ROS-dependent under hypoxia, and HIF-1α mediated SERPINE1 increase induced by ROS via binding to the SERPINE1 promoter region, thereby facilitating glioblastoma migration and invasion. Taken together, our data revealed that hypoxia-induced ROS reinforce the hypoxic adaptation of glioblastoma by driving the HIF-1α-SERPINE1 signaling pathway, and that targeting ROS may be a promising therapeutic strategy for glioblastoma.
Humans
;
Cell Hypoxia
;
Cell Line, Tumor
;
Glioblastoma/pathology*
;
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism*
;
Plasminogen Activator Inhibitor 1/metabolism*
;
Reactive Oxygen Species/metabolism*
;
Signal Transduction
;
Tumor Microenvironment
;
Brain Neoplasms/pathology*
10.Resolving the lineage relationship between malignant cells and vascular cells in glioblastomas.
Fangyu WANG ; Xuan LIU ; Shaowen LI ; Chen ZHAO ; Yumei SUN ; Kuan TIAN ; Junbao WANG ; Wei LI ; Lichao XU ; Jing JING ; Juan WANG ; Sylvia M EVANS ; Zhiqiang LI ; Ying LIU ; Yan ZHOU
Protein & Cell 2023;14(2):105-122
Glioblastoma multiforme (GBM), a highly malignant and heterogeneous brain tumor, contains various types of tumor and non-tumor cells. Whether GBM cells can trans-differentiate into non-neural cell types, including mural cells or endothelial cells (ECs), to support tumor growth and invasion remains controversial. Here we generated two genetic GBM models de novo in immunocompetent mouse brains, mimicking essential pathological and molecular features of human GBMs. Lineage-tracing and transplantation studies demonstrated that, although blood vessels in GBM brains underwent drastic remodeling, evidence of trans-differentiation of GBM cells into vascular cells was barely detected. Intriguingly, GBM cells could promiscuously express markers for mural cells during gliomagenesis. Furthermore, single-cell RNA sequencing showed that patterns of copy number variations (CNVs) of mural cells and ECs were distinct from those of GBM cells, indicating discrete origins of GBM cells and vascular components. Importantly, single-cell CNV analysis of human GBM specimens also suggested that GBM cells and vascular cells are likely separate lineages. Rather than expansion owing to trans-differentiation, vascular cell expanded by proliferation during tumorigenesis. Therefore, cross-lineage trans-differentiation of GBM cells is very unlikely to occur during gliomagenesis. Our findings advance understanding of cell lineage dynamics during gliomagenesis, and have implications for targeted treatment of GBMs.
Mice
;
Animals
;
Humans
;
Glioblastoma/pathology*
;
Endothelial Cells/pathology*
;
DNA Copy Number Variations
;
Brain/metabolism*
;
Brain Neoplasms/pathology*


Result Analysis
Print
Save
E-mail