Treatment of Glioma

A topical collection in Cancers (ISSN 2072-6694). This collection belongs to the section "Cancer Therapy".

Viewed by 38973

Editors


E-Mail Website
Guest Editor
Stamcellscentrum, Lund, Sweden
Interests: glioma

E-Mail Website
Guest Editor
Neurosurgeon at Dept of Neurosurgery, Skåne University Hospital in Lund Rausing Laboratory at Lund University, Lund, Sweden
Interests: glioma

E-Mail Website
Guest Editor
Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
Interests: neurosurgery; glioma; spinal cord injury; neurooncology
Special Issues, Collections and Topics in MDPI journals

Topical Collection Information

Dear Colleagues,

The management and treatment of adult patients with gliomas present some of the most significant challenges in oncology today. The classification of these tumors has recently undergone major changes, which will bear an impact on treatment and patient monitoring. As a result, new guidelines have recently been developed for the diagnosis and treatment of gliomas and several potential practice-changing clinical trials have recently been completed. Entering the era of molecular diagnostics, personalized chemotherapy and immunotherapy, this Special Issue seeks to give an overview of the field.

This Special Issue aims to give an overview of the latest consensus-recommended conventional treatment paradigms as well as to present new investigational clinical treatment modalities for glioma patients. The intention is also to give an overview of brain tumor rehabilitation programs and the patients' own experiences of treatment as reflected in quality of life assessments.

In this Special Issue, original research articles and reviews are welcome. Research areas may include (but are not limited to) the following: radiological and molecular diagnosis and classification, surgical treatment including laser interstitial therapy, chemotherapy, radiotherapy, tumor-treating fields, immunotherapy, novel drug development, drug repurposing, clinical trials, rehabilitation, neuropsychological aspects, cognition and quality of life.

We look forward to receiving your contributions.

Dr. Johan Bengzon
Dr. Henrietta Nittby
Dr. Safwan Alomari
Guest Editors

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 collection 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

  • glioma
  • chemotherapy
  • radiotherapy
  • surgery
  • immunotherapy
  • clinical trials
  • drug development
  • rehabilitation
  • quality of life

Published Papers (15 papers)

2024

Jump to: 2023, 2022

13 pages, 1162 KiB  
Article
Exploring Genetic Determinants: A Comprehensive Analysis of Serpin B Family SNPs and Prognosis in Glioblastoma Multiforme Patients
by Sohaib M. Al-Khatib, Ayah N. Al-Bzour, Mohammad N. Al-Majali, Laila M. Sa’d, Joud A. Alramadneh, Nour R. Othman, Abdel-Hameed Al-Mistarehi and Safwan Alomari
Cancers 2024, 16(6), 1112; https://doi.org/10.3390/cancers16061112 - 10 Mar 2024
Viewed by 737
Abstract
Serpins are serine proteinase inhibitors, with several serpins being overexpressed in cancer cells. Thus, we aim to analyze the single-nucleotide polymorphism (SNP) of Serpinb11 and its association with GBM survival. A cohort of 63 GBM patients recruited from King Abdullah University Hospital in [...] Read more.
Serpins are serine proteinase inhibitors, with several serpins being overexpressed in cancer cells. Thus, we aim to analyze the single-nucleotide polymorphism (SNP) of Serpinb11 and its association with GBM survival. A cohort of 63 GBM patients recruited from King Abdullah University Hospital in Jordan underwent polymorphism analysis and overall survival (OS) assessments. The Cancer Genome Atlas (GBM) cohort was useful for validation. We constructed a risk score using the principal component analysis for the following Serpin genes: Serpinb3, Serpinb5, Serpinb6, Serpinb11, and Serpinb12, and patients were grouped into high- vs. low-risk groups based on the median cutoff. Univariable Cox models were used to study the survival outcomes. We identified a significant association between rs4940595 and survival. In the TCGA cohort, Serpinb3 alterations showed worse OS. Univariable Cox showed worse PFS outcomes with higher SERPINB5 and SERPINB6 expression. A Serpin B 5-gene risk score showed a trend towards worse PFS in the high-risk group. Upregulated DEGs showed GO enrichment in cytokine regulation and production, positive regulation of leukocyte activation, and the MAPK cascade. The high-risk group showed a significantly higher infiltration of M2 macrophages and activated mast cells. Our findings showed a significant role of the Serpin B family in GBM survival in the Jordanian population. Full article
Show Figures

Figure 1

19 pages, 5601 KiB  
Article
Identification of Hypoxia Prognostic Signature in Glioblastoma Multiforme Based on Bulk and Single-Cell RNA-Seq
by Yaman B. Ahmed, Obada E. Ababneh, Anas A. Al-Khalili, Abdullah Serhan, Zaid Hatamleh, Owais Ghammaz, Mohammad Alkhaldi and Safwan Alomari
Cancers 2024, 16(3), 633; https://doi.org/10.3390/cancers16030633 - 01 Feb 2024
Viewed by 1201
Abstract
Glioblastoma (GBM) represents a profoundly aggressive and heterogeneous brain neoplasm linked to a bleak prognosis. Hypoxia, a common feature in GBM, has been linked to tumor progression and therapy resistance. In this study, we aimed to identify hypoxia-related differentially expressed genes (DEGs) and [...] Read more.
Glioblastoma (GBM) represents a profoundly aggressive and heterogeneous brain neoplasm linked to a bleak prognosis. Hypoxia, a common feature in GBM, has been linked to tumor progression and therapy resistance. In this study, we aimed to identify hypoxia-related differentially expressed genes (DEGs) and construct a prognostic signature for GBM patients using multi-omics analysis. Patient cohorts were collected from publicly available databases, including the Gene Expression Omnibus (GEO), the Chinese Glioma Genome Atlas (CGGA), and The Cancer Genome Atlas—Glioblastoma Multiforme (TCGA-GBM), to facilitate a comprehensive analysis. Hypoxia-related genes (HRGs) were obtained from the Molecular Signatures Database (MSigDB). Differential expression analysis revealed 41 hypoxia-related DEGs in GBM patients. A consensus clustering approach, utilizing these DEGs’ expression patterns, identified four distinct clusters, with cluster 1 showing significantly better overall survival. Machine learning techniques, including univariate Cox regression and LASSO regression, delineated a prognostic signature comprising six genes (ANXA1, CALD1, CP, IGFBP2, IGFBP5, and LOX). Multivariate Cox regression analysis substantiated the prognostic significance of a set of three optimal signature genes (CP, IGFBP2, and LOX). Using the hypoxia-related prognostic signature, patients were classified into high- and low-risk categories. Survival analysis demonstrated that the high-risk group exhibited inferior overall survival rates in comparison to the low-risk group. The prognostic signature showed good predictive performance, as indicated by the area under the curve (AUC) values for one-, three-, and five-year overall survival. Furthermore, functional enrichment analysis of the DEGs identified biological processes and pathways associated with hypoxia, providing insights into the underlying mechanisms of GBM. Delving into the tumor immune microenvironment, our analysis revealed correlations relating the hypoxia-related prognostic signature to the infiltration of immune cells in GBM. Overall, our study highlights the potential of a hypoxia-related prognostic signature as a valuable resource for forecasting the survival outcome of GBM patients. The multi-omics approach integrating bulk sequencing, single-cell analysis, and immune microenvironment assessment enhances our understanding of the intricate biology characterizing GBM, thereby potentially informing the tailored design of therapeutic interventions. Full article
Show Figures

Figure 1

2023

Jump to: 2024, 2022

10 pages, 888 KiB  
Article
Unplanned 30-Day Readmission in Glioblastoma Patients: Implications for the Extent of Resection and Adjuvant Therapy
by Johannes Kasper, Johannes Wach, Martin Vychopen, Felix Arlt, Erdem Güresir, Tim Wende and Florian Wilhelmy
Cancers 2023, 15(15), 3907; https://doi.org/10.3390/cancers15153907 - 01 Aug 2023
Cited by 1 | Viewed by 737
Abstract
Background: Unplanned early readmission (UER) within 30 days after hospital release is a negative prognostic marker for patients diagnosed with glioblastoma (GBM). This work analyzes the impact of UER on the effects of standard therapy modalities for GBM patients, including the extent of [...] Read more.
Background: Unplanned early readmission (UER) within 30 days after hospital release is a negative prognostic marker for patients diagnosed with glioblastoma (GBM). This work analyzes the impact of UER on the effects of standard therapy modalities for GBM patients, including the extent of resection (EOR) and adjuvant therapy regimen. Methods: Records were searched for patients with newly diagnosed GBM between 2014 and 2020 who were treated at our facility. Exclusion criteria were being aged below 18 years or missing data. An overall survival (OS) analysis (Kaplan–Meier estimate; Cox regression) was performed on various GBM patient sub-cohorts. Results: A total of 276 patients were included in the study. UER occurred in 13.4% (n = 37) of all cases, significantly reduced median OS (5.7 vs. 14.5 months, p < 0.001 by logrank), and was associated with an increased hazard of mortality (hazard ratio 3.875, p < 0.001) in multivariate Cox regression when other clinical parameters were applied as confounders. The Kaplan–Meier analysis also showed that patients experiencing UER still benefitted from adjuvant radio-chemotherapy when compared to radiotherapy or no adjuvant therapy (p < 0.001 by logrank). A higher EOR did not improve OS in GBM patients with UER (p = 0.659). Conclusion: UER is negatively associated with survival in GBM patients. In contrast to EOR, adjuvant radio-chemotherapy was beneficial, even after UER. Full article
Show Figures

Figure 1

18 pages, 596 KiB  
Review
CAR T Cell Therapy in Glioblastoma: Overcoming Challenges Related to Antigen Expression
by Andrew S. Luksik, Eli Yazigi, Pavan Shah and Christopher M. Jackson
Cancers 2023, 15(5), 1414; https://doi.org/10.3390/cancers15051414 - 23 Feb 2023
Cited by 14 | Viewed by 4121
Abstract
Glioblastoma (GBM) is the most common primary brain tumor, yet prognosis remains dismal with current treatment. Immunotherapeutic strategies have had limited effectiveness to date in GBM, but recent advances hold promise. One such immunotherapeutic advance is chimeric antigen receptor (CAR) T cell therapy, [...] Read more.
Glioblastoma (GBM) is the most common primary brain tumor, yet prognosis remains dismal with current treatment. Immunotherapeutic strategies have had limited effectiveness to date in GBM, but recent advances hold promise. One such immunotherapeutic advance is chimeric antigen receptor (CAR) T cell therapy, where autologous T cells are extracted and engineered to express a specific receptor against a GBM antigen and are then infused back into the patient. There have been numerous preclinical studies showing promising results, and several of these CAR T cell therapies are being tested in clinical trials for GBM and other brain cancers. While results in tumors such as lymphomas and diffuse intrinsic pontine gliomas have been encouraging, early results in GBM have not shown clinical benefit. Potential reasons for this are the limited number of specific antigens in GBM, their heterogenous expression, and their loss after initiating antigen-specific therapy due to immunoediting. Here, we review the current preclinical and clinical experiences with CAR T cell therapy in GBM and potential strategies to develop more effective CAR T cells for this indication. Full article
Show Figures

Figure 1

12 pages, 862 KiB  
Review
CAR-T Therapy in GBM: Current Challenges and Avenues for Improvement
by Ayush Pant and Michael Lim
Cancers 2023, 15(4), 1249; https://doi.org/10.3390/cancers15041249 - 16 Feb 2023
Cited by 8 | Viewed by 2697
Abstract
Completed clinical trials of CAR-T cells in glioblastoma (GBM) have revealed key challenges that limit their efficacy. These include incomplete antigen coverage, downregulation of target antigen in response to therapy, exposure to immunosuppressive cells and cytokines in the tumor microenvironment and exhaustion of [...] Read more.
Completed clinical trials of CAR-T cells in glioblastoma (GBM) have revealed key challenges that limit their efficacy. These include incomplete antigen coverage, downregulation of target antigen in response to therapy, exposure to immunosuppressive cells and cytokines in the tumor microenvironment and exhaustion of CAR-T cells. To overcome these challenges, CAR-T cells have been modified to maximize effector function and resist immunosuppression in the tumor while limiting toxicities to the host. Adoption of these novel CAR-T strategies in GBM can overcome the “cold tumor” phenotype of GBM and trigger an inflammatory cascade that maximizes tumor clearance and minimizes CAR-T dysfunction. To achieve this, understanding and harnessing the antigenic, metabolic and immunological composition of GBM is crucial. Here we review the findings from completed clinical trials of CAR-T cells in GBM as well as novel strategies that could improve CAR-T survival and function in the tumor. Full article
Show Figures

Figure 1

26 pages, 1635 KiB  
Review
Dendrimer Technology in Glioma: Functional Design and Potential Applications
by Hallie Gaitsch, Andrew M. Hersh, Safwan Alomari and Betty M. Tyler
Cancers 2023, 15(4), 1075; https://doi.org/10.3390/cancers15041075 - 08 Feb 2023
Cited by 1 | Viewed by 2225
Abstract
Novel therapeutic and diagnostic methods are sorely needed for gliomas, which contribute yearly to hundreds of thousands of cancer deaths worldwide. Despite the outpouring of research efforts and funding aimed at improving clinical outcomes for patients with glioma, the prognosis for high-grade glioma, [...] Read more.
Novel therapeutic and diagnostic methods are sorely needed for gliomas, which contribute yearly to hundreds of thousands of cancer deaths worldwide. Despite the outpouring of research efforts and funding aimed at improving clinical outcomes for patients with glioma, the prognosis for high-grade glioma, and especially glioblastoma, remains dire. One of the greatest obstacles to improving treatment efficacy and destroying cancer cells is the safe delivery of chemotherapeutic drugs and biologics to the tumor site at a high enough dose to be effective. Over the past few decades, a burst of research has leveraged nanotechnology to overcome this obstacle. There has been a renewed interest in adapting previously understudied dendrimer nanocarriers for this task. Dendrimers are small, highly modifiable, branched structures featuring binding sites for a variety of drugs and ligands. Recent studies have demonstrated the potential for dendrimers and dendrimer conjugates to effectively shuttle therapeutic cargo to the correct tumor location, permeate the tumor, and promote apoptosis of tumor cells while minimizing systemic toxicity and damage to surrounding healthy brain tissue. This review provides a primer on the properties of dendrimers; outlines the mechanisms by which they can target delivery of substances to the site of brain pathology; and delves into current trends in the application of dendrimers to drug and gene delivery, and diagnostic imaging, in glioma. Finally, future directions for translating these in vitro and in vivo findings to the clinic are discussed. Full article
Show Figures

Figure 1

19 pages, 4524 KiB  
Review
Tumor Microenvironment in Gliomas: A Treatment Hurdle or an Opportunity to Grab?
by Vincenzo Di Nunno, Marta Aprile, Lidia Gatto, Alicia Tosoni, Lucia Ranieri, Stefania Bartolini and Enrico Franceschi
Cancers 2023, 15(4), 1042; https://doi.org/10.3390/cancers15041042 - 07 Feb 2023
Cited by 4 | Viewed by 2823
Abstract
Gliomas are the most frequent central nervous system (CNS) primary tumors. The prognosis and clinical outcomes of these malignancies strongly diverge according to their molecular alterations and range from a few months to decades. The tumor-associated microenvironment involves all cells and connective tissues [...] Read more.
Gliomas are the most frequent central nervous system (CNS) primary tumors. The prognosis and clinical outcomes of these malignancies strongly diverge according to their molecular alterations and range from a few months to decades. The tumor-associated microenvironment involves all cells and connective tissues surrounding tumor cells. The composition of the microenvironment as well as the interactions with associated neoplastic mass, are both variables assuming an increasing interest in these last years. This is mainly because the microenvironment can mediate progression, invasion, dedifferentiation, resistance to treatment, and relapse of primary gliomas. In particular, the tumor microenvironment strongly diverges from isocitrate dehydrogenase (IDH) mutated and wild-type (wt) tumors. Indeed, IDH mutated gliomas often show a lower infiltration of immune cells with reduced angiogenesis as compared to IDH wt gliomas. On the other hand, IDH wt tumors exhibit a strong immune infiltration mediated by several cytokines and chemokines, including CCL2, CCL7, GDNF, CSF-1, GM-CSF, etc. The presence of several factors, including Sox2, Oct4, PD-L1, FAS-L, and TGF β2, also mediate an immune switch toward a regulatory inhibited immune system. Other important interactions are described between IDH wt glioblastoma cells and astrocytes, neurons, and stem cells, while these interactions are less elucidated in IDH-mutated tumors. The possibility of targeting the microenvironment is an intriguing perspective in terms of therapeutic drug development. In this review, we summarized available evidence related to the glioma microenvironment, focusing on differences within different glioma subtypes and on possible therapeutic development. Full article
Show Figures

Figure 1

2022

Jump to: 2024, 2023

23 pages, 2139 KiB  
Review
Investigational Microbiological Therapy for Glioma
by Jing Wang, Yaxue Liu, Aohan Zhang, Wenxin Yu, Qian Lei, Bo Xiao and Zhaohui Luo
Cancers 2022, 14(23), 5977; https://doi.org/10.3390/cancers14235977 - 02 Dec 2022
Cited by 1 | Viewed by 2223
Abstract
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median [...] Read more.
Glioma is the most common primary malignancy of the central nervous system (CNS), and 50% of patients present with glioblastoma (GBM), which is the most aggressive type. Currently, the most popular therapies are progressive chemotherapy and treatment with temozolomide (TMZ), but the median survival of glioma patients is still low as a result of the emergence of drug resistance, so we urgently need to find new therapies. A growing number of studies have shown that the diversity, bioactivity, and manipulability of microorganisms make microbial therapy a promising approach for cancer treatment. However, the many studies on the research progress of microorganisms and their derivatives in the development and treatment of glioma are scattered, and nobody has yet provided a comprehensive summary of them. Therefore, in this paper, we review the research progress of microorganisms and their derivatives in the development and treatment of glioma and conclude that it is possible to treat glioma by exogenous microbial therapies and targeting the gut–brain axis. In this article, we discuss the prospects and pressing issues relating to these therapies with the aim of providing new ideas for the treatment of glioma. Full article
Show Figures

Figure 1

22 pages, 661 KiB  
Review
Review of Intraoperative Adjuncts for Maximal Safe Resection of Gliomas and Its Impact on Outcomes
by Hani Chanbour and Silky Chotai
Cancers 2022, 14(22), 5705; https://doi.org/10.3390/cancers14225705 - 21 Nov 2022
Cited by 5 | Viewed by 1888
Abstract
Maximal safe resection is the mainstay of treatment in the neurosurgical management of gliomas, and preserving functional integrity is linked to favorable outcomes. How these modalities differ in their effectiveness on the extent of resection (EOR), survival, and complications remains unknown. A systematic [...] Read more.
Maximal safe resection is the mainstay of treatment in the neurosurgical management of gliomas, and preserving functional integrity is linked to favorable outcomes. How these modalities differ in their effectiveness on the extent of resection (EOR), survival, and complications remains unknown. A systematic literature search was performed with the following inclusion criteria: published between 2005 and 2022, involving brain glioma surgery, and including one or a combination of intraoperative modalities: intraoperative magnetic resonance imaging (iMRI), awake/general anesthesia craniotomy mapping (AC/GA), fluorescence-guided imaging, or combined modalities. Of 525 articles, 464 were excluded and 61 articles were included, involving 5221 glioma patients, 7(11.4%) articles used iMRI, 21(36.8%) used cortical mapping, 15(24.5%) used 5-aminolevulinic acid (5-ALA) or fluorescein sodium, and 18(29.5%) used combined modalities. The heterogeneity in reporting the amount of surgical resection prevented further analysis. Progression-free survival/overall survival (PFS/OS) were reported in 18/61(29.5%) articles, while complications and permanent disability were reported in 38/61(62.2%) articles. The reviewed studies demonstrate that intraoperative adjuncts such as iMRI, AC/GA mapping, fluorescence-guided imaging, and a combination of these modalities improve EOR. However, PFS/OS were underreported. Combining multiple intraoperative modalities seems to have the highest effect compared to each adjunct alone. Full article
Show Figures

Figure 1

31 pages, 1178 KiB  
Review
Smart Nanoformulations for Brain Cancer Theranostics: Challenges and Promises
by Faraz Ahmad, Ressin Varghese, Subhrajita Panda, Siva Ramamoorthy, Mohammad Y. Areeshi, Sharmila Fagoonee and Shafiul Haque
Cancers 2022, 14(21), 5389; https://doi.org/10.3390/cancers14215389 - 01 Nov 2022
Cited by 7 | Viewed by 2708
Abstract
Despite their low prevalence, brain tumors are among the most lethal cancers. They are extremely difficult to diagnose, monitor and treat. Conventional anti-cancer strategies such as radio- and chemotherapy have largely failed, and to date, the development of even a single effective therapeutic [...] Read more.
Despite their low prevalence, brain tumors are among the most lethal cancers. They are extremely difficult to diagnose, monitor and treat. Conventional anti-cancer strategies such as radio- and chemotherapy have largely failed, and to date, the development of even a single effective therapeutic strategy against central nervous system (CNS) tumors has remained elusive. There are several factors responsible for this. Brain cancers are a heterogeneous group of diseases with variable origins, biochemical properties and degrees of invasiveness. High-grade gliomas are amongst the most metastatic and invasive cancers, which is another reason for therapeutic failure in their case. Moreover, crossing the blood brain and the blood brain tumor barriers has been a significant hindrance in the development of efficient CNS therapeutics. Cancer nanomedicine, which encompasses the application of nanotechnology for diagnosis, monitoring and therapy of cancers, is a rapidly evolving field of translational medicine. Nanoformulations, because of their extreme versatility and manipulative potential, are emerging candidates for tumor targeting, penetration and treatment in the brain. Moreover, suitable nanocarriers can be commissioned for theranostics, a combinatorial personalized approach for simultaneous imaging and therapy. This review first details the recent advances in novel bioengineering techniques that provide promising avenues for circumventing the hurdles of delivering the diagnostic/therapeutic agent to the CNS. The authors then describe in detail the tremendous potential of utilizing nanotechnology, particularly nano-theranostics for brain cancer imaging and therapy, and outline the different categories of recently developed next-generation smart nanoformulations that have exceptional potential for making a breakthrough in clinical neuro-oncology therapeutics. Full article
Show Figures

Figure 1

26 pages, 1391 KiB  
Review
Applications of Focused Ultrasound for the Treatment of Glioblastoma: A New Frontier
by Andrew M. Hersh, Meghana Bhimreddy, Carly Weber-Levine, Kelly Jiang, Safwan Alomari, Nicholas Theodore, Amir Manbachi and Betty M. Tyler
Cancers 2022, 14(19), 4920; https://doi.org/10.3390/cancers14194920 - 08 Oct 2022
Cited by 20 | Viewed by 4453
Abstract
Glioblastoma (GBM) is an aggressive primary astrocytoma associated with short overall survival. Treatment for GBM primarily consists of maximal safe surgical resection, radiation therapy, and chemotherapy using temozolomide. Nonetheless, recurrence and tumor progression is the norm, driven by tumor stem cell activity and [...] Read more.
Glioblastoma (GBM) is an aggressive primary astrocytoma associated with short overall survival. Treatment for GBM primarily consists of maximal safe surgical resection, radiation therapy, and chemotherapy using temozolomide. Nonetheless, recurrence and tumor progression is the norm, driven by tumor stem cell activity and a high mutational burden. Focused ultrasound (FUS) has shown promising results in preclinical and clinical trials for treatment of GBM and has received regulatory approval for the treatment of other neoplasms. Here, we review the range of applications for FUS in the treatment of GBM, which depend on parameters, including frequency, power, pulse duration, and duty cycle. Low-intensity FUS can be used to transiently open the blood–brain barrier (BBB), which restricts diffusion of most macromolecules and therapeutic agents into the brain. Under guidance from magnetic resonance imaging, the BBB can be targeted in a precise location to permit diffusion of molecules only at the vicinity of the tumor, preventing side effects to healthy tissue. BBB opening can also be used to improve detection of cell-free tumor DNA with liquid biopsies, allowing non-invasive diagnosis and identification of molecular mutations. High-intensity FUS can cause tumor ablation via a hyperthermic effect. Additionally, FUS can stimulate immunological attack of tumor cells, can activate sonosensitizers to exert cytotoxic effects on tumor tissue, and can sensitize tumors to radiation therapy. Finally, another mechanism under investigation, known as histotripsy, produces tumor ablation via acoustic cavitation rather than thermal effects. Full article
Show Figures

Figure 1

34 pages, 1712 KiB  
Review
Molecular Pathways and Genomic Landscape of Glioblastoma Stem Cells: Opportunities for Targeted Therapy
by Andrew M. Hersh, Hallie Gaitsch, Safwan Alomari, Daniel Lubelski and Betty M. Tyler
Cancers 2022, 14(15), 3743; https://doi.org/10.3390/cancers14153743 - 31 Jul 2022
Cited by 14 | Viewed by 4016
Abstract
Glioblastoma (GBM) is an aggressive tumor of the central nervous system categorized by the World Health Organization as a Grade 4 astrocytoma. Despite treatment with surgical resection, adjuvant chemotherapy, and radiation therapy, outcomes remain poor, with a median survival of only 14-16 months. [...] Read more.
Glioblastoma (GBM) is an aggressive tumor of the central nervous system categorized by the World Health Organization as a Grade 4 astrocytoma. Despite treatment with surgical resection, adjuvant chemotherapy, and radiation therapy, outcomes remain poor, with a median survival of only 14-16 months. Although tumor regression is often observed initially after treatment, long-term recurrence or progression invariably occurs. Tumor growth, invasion, and recurrence is mediated by a unique population of glioblastoma stem cells (GSCs). Their high mutation rate and dysregulated transcriptional landscape augment their resistance to conventional chemotherapy and radiation therapy, explaining the poor outcomes observed in patients. Consequently, GSCs have emerged as targets of interest in new treatment paradigms. Here, we review the unique properties of GSCs, including their interactions with the hypoxic microenvironment that drives their proliferation. We discuss vital signaling pathways in GSCs that mediate stemness, self-renewal, proliferation, and invasion, including the Notch, epidermal growth factor receptor, phosphatidylinositol 3-kinase/Akt, sonic hedgehog, transforming growth factor beta, Wnt, signal transducer and activator of transcription 3, and inhibitors of differentiation pathways. We also review epigenomic changes in GSCs that influence their transcriptional state, including DNA methylation, histone methylation and acetylation, and miRNA expression. The constituent molecular components of the signaling pathways and epigenomic regulators represent potential sites for targeted therapy, and representative examples of inhibitory molecules and pharmaceuticals are discussed. Continued investigation into the molecular pathways of GSCs and candidate therapeutics is needed to discover new effective treatments for GBM and improve survival. Full article
Show Figures

Figure 1

14 pages, 1525 KiB  
Article
Identification of a Prognostic Microenvironment-Related Gene Signature in Glioblastoma Patients Treated with Carmustine Wafers
by Ivana Manini, Emiliano Dalla, Vera Vendramin, Daniela Cesselli, Carla Di Loreto, Miran Skrap and Tamara Ius
Cancers 2022, 14(14), 3413; https://doi.org/10.3390/cancers14143413 - 14 Jul 2022
Cited by 1 | Viewed by 1646
Abstract
Despite the state-of-the-art treatment, patients diagnosed with glioblastoma (GBM) have a median overall survival (OS) of 14 months. The insertion of carmustine wafers (CWs) into the resection cavity as adjuvant treatment represents a promising option, although its use has been limited due to [...] Read more.
Despite the state-of-the-art treatment, patients diagnosed with glioblastoma (GBM) have a median overall survival (OS) of 14 months. The insertion of carmustine wafers (CWs) into the resection cavity as adjuvant treatment represents a promising option, although its use has been limited due to contrasting clinical results. Our retrospective evaluation of CW efficacy showed a significant improvement in terms of OS in a subgroup of patients. Given the crucial role of the tumor microenvironment (TME) in GBM progression and response to therapy, we hypothesized that the TME of patients who benefited from CW could have different properties compared to that of patients who did not show any advantage. Using an in vitro model of the glioma microenvironment, represented by glioma-associated-stem cells (GASC), we performed a transcriptomic analysis of GASC isolated from tumors of patients responsive and not responsive to CW to identify differentially expressed genes. We found different transcriptomic profiles, and we identified four genes, specifically down-regulated in GASC isolated from long-term survivors, correlated with clinical data deposited in the TCGA–GBM dataset. Our results highlight that studying the in vitro properties of patient-specific glioma microenvironments can help to identify molecular determinants potentially prognostic for patients treated with CW. Full article
Show Figures

Figure 1

21 pages, 1242 KiB  
Article
A Potential New Treatment for High-Grade Glioma: A Study Assessing Repurposed Drug Combinations against Patient-Derived High-Grade Glioma Cells
by Sarah Lastakchi, Mary Kanyinsola Olaloko and Christopher McConville
Cancers 2022, 14(11), 2602; https://doi.org/10.3390/cancers14112602 - 25 May 2022
Cited by 3 | Viewed by 2323
Abstract
Repurposed drugs have demonstrated in vitro success against high-grade gliomas; however, their clinical success has been limited due to the in vitro model not truly representing the clinical scenario. In this study, we used two distinct patient-derived tumour fragments (tumour core (TC) and [...] Read more.
Repurposed drugs have demonstrated in vitro success against high-grade gliomas; however, their clinical success has been limited due to the in vitro model not truly representing the clinical scenario. In this study, we used two distinct patient-derived tumour fragments (tumour core (TC) and tumour margin (TM)) to generate a heterogeneous, clinically relevant in vitro model to assess if a combination of repurposed drugs (irinotecan, pitavastatin, disulfiram, copper gluconate, captopril, celecoxib, itraconazole and ticlopidine), each targeting a different growth promoting pathway, could successfully treat high-grade gliomas. To ensure the clinical relevance of our data, TC and TM samples from 11 different patients were utilized. Our data demonstrate that, at a concentration of 100µm or lower, all drug combinations achieved lower LogIC50 values than temozolomide, with one of the combinations almost eradicating the cancer by achieving cell viabilities below 4% in five of the TM samples 6 days after treatment. Temozolomide was unable to stop tumour growth over the 14-day assay, while combination 1 stopped tumour growth, with combinations 2, 3 and 4 slowing down tumour growth at higher doses. To validate the cytotoxicity data, we used two distinct assays, end point MTT and real-time IncuCyte life analysis, to evaluate the cytotoxicity of the combinations on the TC fragment from patient 3, with the cell viabilities comparable across both assays. The local administration of combinations of repurposed drugs that target different growth promoting pathways of high-grade gliomas have the potential to be translated into the clinic as a novel treatment strategy for high-grade gliomas. Full article
Show Figures

Figure 1

15 pages, 912 KiB  
Systematic Review
Gliomas Infiltrating the Corpus Callosum: A Systematic Review of the Literature
by Paolo Palmisciano, Gianluca Ferini, Gina Watanabe, Christian Ogasawara, Emal Lesha, Othman Bin-Alamer, Giuseppe E. Umana, Kenny Yu, Aaron A. Cohen-Gadol, Tarek Y. El Ahmadieh and Ali S. Haider
Cancers 2022, 14(10), 2507; https://doi.org/10.3390/cancers14102507 - 19 May 2022
Cited by 6 | Viewed by 3385
Abstract
Background: Gliomas infiltrating the corpus callosum (G-I-CC) majorly impact patient quality-of-life, but maximally safe tumor resection is challenging. We systematically reviewed the literature on G-I-CC. Methods: PubMed, EMBASE, Scopus, Web of Science, and Cochrane were searched following the PRISMA guidelines to include studies [...] Read more.
Background: Gliomas infiltrating the corpus callosum (G-I-CC) majorly impact patient quality-of-life, but maximally safe tumor resection is challenging. We systematically reviewed the literature on G-I-CC. Methods: PubMed, EMBASE, Scopus, Web of Science, and Cochrane were searched following the PRISMA guidelines to include studies of patients with G-I-CC. Clinicopathological features, treatments, and outcomes were analyzed. Results: We included 52 studies comprising 683 patients. Most patients experienced headache (33%), cognitive decline (18.7%), and seizures (17.7%). Tumors mostly infiltrated the corpus callosum genu (44.2%) with bilateral extension (85.4%) into frontal (68.3%) or parietal (8.9%) lobes. Most G-I-CC were glioblastomas (84.5%) with IDH-wildtype (84.9%) and unmethylated MGMT promoter (53.5%). Resection (76.7%) was preferred over biopsy (23.3%), mostly gross-total (33.8%) and subtotal (32.5%). The tumor-infiltrated corpus callosum was resected in 57.8% of cases. Radiation was delivered in 65.8% of patients and temozolomide in 68.3%. Median follow-up was 12 months (range, 0.1–116). In total, 142 patients (31.8%) experienced post-surgical complications, including transient supplementary motor area syndrome (5.1%) and persistent motor deficits (4.3%) or abulia (2.5%). Post-treatment symptom improvement was reported in 42.9% of patients. No differences in rates of complications (p = 0.231) and symptom improvement (p = 0.375) were found in cases with resected versus preserved corpus callosum. Recurrences occurred in 40.9% of cases, with median progression-free survival of 9 months (0.1–72). Median overall survival was 10.7 months (range, 0.1–116), significantly longer in low-grade tumors (p = 0.013) and after resection (p < 0.001), especially gross-total (p = 0.041) in patients with high-grade tumors. Conclusions: G-I-CC show clinicopathological patterns comparable to other more frequent gliomas. Maximally safe resection significantly improves survival with low rates of persistent complications. Full article
Show Figures

Figure 1

Back to TopTop