Glioblastoma: Recent Advances and Challenges

A special issue of Cancers (ISSN 2072-6694). This special issue belongs to the section "Cancer Therapy".

Deadline for manuscript submissions: 30 September 2024 | Viewed by 5437

Special Issue Editor


E-Mail Website
Guest Editor
UCLA Neuro-Oncology Program, Department of Neurology, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
Interests: brain cancer; gliomas; neurofibromatosis

Special Issue Information

Dear Colleagues,

Glioblastoma (GBM) is the most common and most malignant of all primary tumors of the brain and nervous system. To date, treatments with standard radiation and chemotherapy have provided little additional survival benefits to patients with GBM. Unlike other cancers, there are limited FDA-approved treatments for GBM, and different treatment modalities with proven success in preclinical settings have not been successfully translated to the clinic.Overall, more treatments options are critically needed specifically to target GBM. Various treatment options for GBM that have been tested still have significant limitations regarding brain tumor penetration and the heterogeneity of this cancer. New cancer therapeutics such as immunotherapies are still being evaluated for the treatment of GBM. Better understanding in GBM cancer biology and tumor microenvironment would considerably improve new treatments for GBM.

In this Special Issue, advances in the treatment of GBM will highlight needed areas of studies to enhance treatment options for patients with GBM from a basic and clinical standpoint. 

Dr. P. Leia Nghiemphu
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cancers is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • glioblastoma
  • cancer immunotherapy
  • brain tumor microenvironment
  • molecular targeted therapy
  • GBM

Published Papers (3 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

20 pages, 11248 KiB  
Article
Novel Function of Cancer Stem Cell Marker ALDH1A3 in Glioblastoma: Pro-Angiogenesis through Paracrine PAI-1 and IL-8
by Zhen Chen, Rainer Will, Su Na Kim, Maike Anna Busch, Nicole Dünker, Philipp Dammann, Ulrich Sure and Yuan Zhu
Cancers 2023, 15(17), 4422; https://doi.org/10.3390/cancers15174422 - 04 Sep 2023
Cited by 2 | Viewed by 1324
Abstract
Hyper-angiogenesis is a typical feature of glioblastoma (GBM), the most aggressive brain tumor. We have reported the expression of aldehyde dehydrogenase 1A3 (ALDH1A3) in proliferating vasculature in GBM patients. We hypothesized that ALDH1A3 may act as an angiogenesis promoter in GBM. Two GBM [...] Read more.
Hyper-angiogenesis is a typical feature of glioblastoma (GBM), the most aggressive brain tumor. We have reported the expression of aldehyde dehydrogenase 1A3 (ALDH1A3) in proliferating vasculature in GBM patients. We hypothesized that ALDH1A3 may act as an angiogenesis promoter in GBM. Two GBM cell lines were lentivirally transduced with either ALDH1A3 (ox) or an empty vector (ev). The angiogenesis phenotype was studied in indirect and direct co-culture of endothelial cells (ECs) with oxGBM cells (oxGBMs) and in an angiogenesis model in vivo. Angiogenesis array was performed in oxGBMs. RT2-PCR, Western blot, and double-immunofluorescence staining were performed to confirm the expression of targets identified from the array. A significantly activated angiogenesis phenotype was observed in ECs indirectly and directly co-cultured with oxGBMs and in vivo. Overexpression of ALDH1A3 (oxALDH1A3) led to a marked upregulation of PAI-1 and IL-8 mRNA and protein and a consequential increased release of both proteins. Moreover, oxALDH1A3-induced angiogenesis was abolished by the treatment of the specific inhibitors, respectively, of PAI-1 and IL-8 receptors, CXCR1/2. This study defined ALDH1A3 as a novel angiogenesis promoter. oxALDH1A3 in GBM cells stimulated EC angiogenesis via paracrine upregulation of PAI-1 and IL-8, suggesting ALDH1A3-PAI-1/IL-8 as a novel signaling for future anti-angiogenesis therapy in GBM. Full article
(This article belongs to the Special Issue Glioblastoma: Recent Advances and Challenges)
Show Figures

Figure 1

15 pages, 8837 KiB  
Article
Vasculogenic Mimicry Occurs at Low Levels in Primary and Recurrent Glioblastoma
by Kelsey Maddison, Sam Faulkner, Moira C. Graves, Michael Fay, Nikola A. Bowden and Paul A. Tooney
Cancers 2023, 15(15), 3922; https://doi.org/10.3390/cancers15153922 - 01 Aug 2023
Viewed by 1094
Abstract
Vasculogenic mimicry (VM), the ability of tumour cells to form functional microvasculature without an endothelial lining, may contribute to anti-angiogenic treatment resistance in glioblastoma. We aimed to assess the extent of VM formation in primary and recurrent glioblastomas and to determine whether VM [...] Read more.
Vasculogenic mimicry (VM), the ability of tumour cells to form functional microvasculature without an endothelial lining, may contribute to anti-angiogenic treatment resistance in glioblastoma. We aimed to assess the extent of VM formation in primary and recurrent glioblastomas and to determine whether VM vessels also express prostate-specific membrane antigen (PSMA), a pathological vessel marker. Formalin-fixed paraffin-embedded tissue from 35 matched pairs of primary and recurrent glioblastoma was immunohistochemically labelled for PSMA and CD34 and stained with periodic acid–Schiff (PAS). Vascular structures were categorised as endothelial vessels (CD34+/PAS+) or VM (CD34−/PAS+). Most blood vessels in both primary and recurrent tumours were endothelial vessels, and these significantly decreased in recurrent tumours (p < 0.001). PSMA was expressed by endothelial vessels, and its expression was also decreased in recurrent tumours (p = 0.027). VM was observed in 42.86% of primary tumours and 28.57% of recurrent tumours. VM accounted for only a small proportion of the tumour vasculature and VM density did not differ between primary and recurrent tumours (p = 0.266). The functional contribution of VM and its potential as a treatment target in glioblastoma require further investigation. Full article
(This article belongs to the Special Issue Glioblastoma: Recent Advances and Challenges)
Show Figures

Figure 1

14 pages, 2481 KiB  
Article
Anti-Vimentin Nanobody Decreases Glioblastoma Cell Invasion In Vitro and In Vivo
by Alja Zottel, Metka Novak, Neja Šamec, Bernarda Majc, Sara Colja, Mojca Katrašnik, Miloš Vittori, Barbara Hrastar, Ana Rotter, Andrej Porčnik, Tamara Lah Turnšek, Radovan Komel, Barbara Breznik and Ivana Jovčevska
Cancers 2023, 15(3), 573; https://doi.org/10.3390/cancers15030573 - 17 Jan 2023
Cited by 4 | Viewed by 2157
Abstract
Purpose: Glioblastoma (GBM) is the most common primary brain tumour and one of the deadliest cancers. In addition to late diagnosis and inadequate treatment, the extremely low survival rate is also due to the lack of appropriate therapeutic biomarkers and corresponding therapeutic agents. [...] Read more.
Purpose: Glioblastoma (GBM) is the most common primary brain tumour and one of the deadliest cancers. In addition to late diagnosis and inadequate treatment, the extremely low survival rate is also due to the lack of appropriate therapeutic biomarkers and corresponding therapeutic agents. One of the potential therapeutic biomarkers is the intermediate filament vimentin, which is associated with epithelial-mesenchymal transition (EMT). The purpose of this study was to analyse the effect of the anti-vimentin nanobody Nb79 on cell invasion in vitro and in vivo. To further our understanding of the mechanism of action, we investigated the association between Nb79 and EMT in GBM and GBM stem cells by analysing the expression levels of key EMT-related proteins. Methods: The expression of vimentin in glioma tissues and cells was determined by RT-qPCR. An invasion assay was performed on differentiated glioblastoma cell line U-87 MG and stem cell line NCH421k in vitro as well as in vivo in zebrafish embryos. The effect of Nb79 on expression of EMT biomarkers beta-catenin, vimentin, ZEB-1 and ZO1 was determined by Western blot and immunocytochemistry. Results: Our study shows that vimentin is upregulated in glioblastoma tissue compared to lower grade glioma and non-tumour brain tissue. We demonstrated that treatment with Nb79 reduced glioblastoma cell invasion by up to 64% in vitro and up to 21% in vivo. In addition, we found that the tight junction protein ZO-1 had higher expression on the cell membrane, when treated with inhibitory anti-vimentin Nb79 compared to control. Conclusion: In conclusion, our results suggest that anti-vimentin nanobody Nb79 is a promising tool to target glioblastoma cell invasion. Full article
(This article belongs to the Special Issue Glioblastoma: Recent Advances and Challenges)
Show Figures

Figure 1

Planned Papers

The below list represents only planned manuscripts. Some of these manuscripts have not been received by the Editorial Office yet. Papers submitted to MDPI journals are subject to peer-review.

Title: Glioblastoma Multiforme Therapy: Present and Future
Authors: María A. Marqués-Torrejón1*, Paz Moreno-Murciano2, María Oriol-Caballo2,3, Rafael López-Blanch2,3, Alba Loras1, Luis G. Gonzalez-Bonet4, Conrado Martinez-Cadenas5, José M. Estrela2,3,6, and Elena Obra
Affiliation: 3Department of Medicine, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain 2 Scientia BioTech S.L., 46002 Valencia, Spain 1 Cell Pathophysiology Unit (UFC), Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain 4 Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain 5 Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain 6 Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain *Correspondence: torrejom@uji.es M.A.M.-T.), Tel.: +34-641774524; elena.obrador@uv.es (E.O.), Tel.: +34-963864646
Abstract: The glioblastoma multiforme (GBM, also called grade IV astrocytoma) is the most aggressive and prevalent brain tumor that forms from astrocytes of the brain and spinal cord. It is more frequent in adults and affects the brain more than the spinal cord. The standard of care for newly diagnosed glioblastoma multiforme (GBM) involves surgical resection (if feasible), concurrent radiotherapy and chemotherapy (temozolomide seems to work better by sensitizing the GBM cells to radiation), targeted therapy in recurrent GBM (e.g. bevacizumab, which selectively binds to the VEGF and reduces the growth of blood vessels that supply the tumor with nutrients and oxygen), with or without tumor treating fields (mild electrical fields that pulse through the skin of the scalp aiming to interrupt cancer cells' ability to divide). Despite multimodality treatment, recurrence is almost universal with a median survival of <2 years. Ongoing research, aiming to change this grim prognosis, includes targeted therapies interfering at different signaling mechanisms (PARP, CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RTK/RAS/RAF/MEK), immunotherapies (checkpoint inhibitors, vaccines, CAR T cells, NK cell-based therapy), oncolytic virotherapy (specific genes are delivered into target cells with viral vectors to induce oncolysis and the host immune response), nanotherapies (nanometric structures having active anti-GBM agents such as immune cells, chemotherapeutic/anti-angiogenic drugs or sensitizers, or molecules which target GBM cellular receptors/angiogenic blood vessels or open the BBB), and treatments based on the use of non-ionizing energies (laser interstitial thermal therapy, high intensity focused ultrasounds and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies, and to present novel approaches which are under development or following clinical trials. Keywords: glioblastoma multiforme, targeted therapies, immunotherapies, viral therapies, nanotherapies, non-ionizing radiations

Back to TopTop