ijms-logo

Journal Browser

Journal Browser

Frontiers in Neuro-Oncology

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Oncology".

Deadline for manuscript submissions: closed (31 July 2022) | Viewed by 57670

Special Issue Editor

Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
Interests: angiogenesis; blood-brain barrier; blood-tumor barrier; brain cancer treatment; brain metastasis; intercellular communication; glioma; tumor microenvironment; tumor heterogeneity; tumor pericytes
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The majority of central nervous system neoplasms are brain tumors, with estimates indicating that around 77% of primary brain tumors (with origin in brain cells) are gliomas and that secondary brain tumors (metastases) are approximately three times more common than primary ones. Brain tumors are life-threating conditions which account for the complexity of the disease, involving the hijack of brain and immune cells for tumor growth and the activation of compensatory signaling mechanisms in malignant cells that lead to therapeutic resistance. This Special Issue intends to gather forefront knowledge about brain neoplasms, including the signaling pathways involved in malignant cell survival, proliferation and invasion, tumor microenvironment and intercellular communication, angiogenesis, and vascular co-option, as well as inter- and intra-tumoral heterogeneity. It further aims to highlight novel therapeutic strategies with the ability to circumvent the blood–brain barrier and blood–tumor barrier and to specifically target malignant cells, as well as innovative study and imaging tools.

By bringing together state-of-the-art concepts on relevant topics, this Special Issue shall contribute towards a better understanding of brain tumor biology and diversity, essential for the identification of targets and development of novel and personalized therapeutics to improve patients’ life quality and expectancy.

Prof. Maria Alexandra Brito
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. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. 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

  • Angiogenesis
  • Blood-brain barrier
  • Blood-tumor barrier
  • Brain cancer treatment
  • Brain metastasis
  • Intercellular communication
  • Glioma
  • Tumor microenvironment
  • Tumor heterogeneity
  • Tumor pericytes

Published Papers (21 papers)

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

Research

Jump to: Review

20 pages, 3401 KiB  
Article
Metabolic Heterogeneity of Brain Tumor Cells of Proneural and Mesenchymal Origin
by Corinna Seliger, Anne-Louise Meyer, Verena Leidgens, Lisa Rauer, Sylvia Moeckel, Birgit Jachnik, Judith Proske, Katja Dettmer, Tanja Rothhammer-Hampl, Leon D. Kaulen, Markus J. Riemenschneider, Peter J. Oefner, Marina Kreutz, Nils-Ole Schmidt, Marsha Merrill, Martin Uhl, Kathrin Renner, Arabel Vollmann-Zwerenz, Martin Proescholdt and Peter Hau
Int. J. Mol. Sci. 2022, 23(19), 11629; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911629 - 01 Oct 2022
Cited by 5 | Viewed by 1668
Abstract
Brain-tumor-initiating cells (BTICs) of proneural and mesenchymal origin contribute to the highly malignant phenotype of glioblastoma (GB) and resistance to current therapies. BTICs of different subtypes were challenged with oxidative phosphorylation (OXPHOS) inhibition with metformin to assess the differential effects of metabolic intervention [...] Read more.
Brain-tumor-initiating cells (BTICs) of proneural and mesenchymal origin contribute to the highly malignant phenotype of glioblastoma (GB) and resistance to current therapies. BTICs of different subtypes were challenged with oxidative phosphorylation (OXPHOS) inhibition with metformin to assess the differential effects of metabolic intervention on key resistance features. Whereas mesenchymal BTICs varied according to their invasiveness, they were in general more glycolytic and less responsive to metformin. Proneural BTICs were less invasive, catabolized glucose more via the pentose phosphate pathway, and responded better to metformin. Targeting glycolysis may be a promising approach to inhibit tumor cells of mesenchymal origin, whereas proneural cells are more responsive to OXPHOS inhibition. Future clinical trials exploring metabolic interventions should account for metabolic heterogeneity of brain tumors. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

18 pages, 4030 KiB  
Article
LPPR5 Expression in Glioma Affects Growth, Vascular Architecture, and Sunitinib Resistance
by Lena Stange, Kristin Elizabeth Lucia, Adnan Ghori, Peter Vajkoczy, Marcus Czabanka and Thomas Broggini
Int. J. Mol. Sci. 2022, 23(6), 3108; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23063108 - 13 Mar 2022
Cited by 3 | Viewed by 2470
Abstract
Despite intensive research, glioblastoma remains almost invariably fatal. Various promising drugs targeting specific aspects of glioma biology, in addition to or as an alternative to antiproliferative chemotherapy, were not successful in larger clinical trials. Further insights into the biology of glioma and the [...] Read more.
Despite intensive research, glioblastoma remains almost invariably fatal. Various promising drugs targeting specific aspects of glioma biology, in addition to or as an alternative to antiproliferative chemotherapy, were not successful in larger clinical trials. Further insights into the biology of glioma and the mechanisms behind the evasive-adaptive response to targeted therapies is needed to help identify new therapeutic targets, prognostics, or predictive biomarkers. As a modulator of the canonically oncogenic Rho-GTPase pathway, Lipid phosphate phosphatase-related protein type 5 (LPPR5) is pivotal in influencing growth, angiogenesis, and therapeutic resistance. We used a GL261 murine orthotopic allograft glioma model to quantify the tumor growth and to obtain tissue for histological and molecular analysis. Epicortical intravital epi-illumination fluorescence video microscopy of the tumor cell spheroids was used to characterize the neovascular architecture and hemodynamics. GL261-glioma growth was delayed and decelerated after LPPR5 overexpression (LPPR5OE). We observed increased tumor cell apoptosis and decreased expression and secretion of vascular endothelial growth factor A in LPPR5OE glioma. Hence, an altered micro-angioarchitecture consisting of dysfunctional small blood vessels was discovered in the LPPR5OE tumors. Sunitinib therapy eliminated these vessels but had no effect on tumor growth or apoptosis. In general, LPPR5 overexpression generated a more benign, proapoptotic glioma phenotype with delayed growth and a dysfunctional vascular architecture. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

18 pages, 2683 KiB  
Article
Genome-Wide Expression and Anti-Proliferative Effects of Electric Field Therapy on Pediatric and Adult Brain Tumors
by Joshua Branter, Maria Estevez-Cebrero, Mohammed Diksin, Michaela Griffin, Marcos Castellanos-Uribe, Sean May, Ruman Rahman, Richard Grundy, Surajit Basu and Stuart Smith
Int. J. Mol. Sci. 2022, 23(4), 1982; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23041982 - 11 Feb 2022
Cited by 7 | Viewed by 2399
Abstract
The lack of treatment options for high-grade brain tumors has led to searches for alternative therapeutic modalities. Electrical field therapy is one such area. The Optune™ system is an FDA-approved novel device that delivers continuous alternating electric fields (tumor treating fields—TTFields) to the [...] Read more.
The lack of treatment options for high-grade brain tumors has led to searches for alternative therapeutic modalities. Electrical field therapy is one such area. The Optune™ system is an FDA-approved novel device that delivers continuous alternating electric fields (tumor treating fields—TTFields) to the patient for the treatment of primary and recurrent Glioblastoma multiforme (GBM). Various mechanisms have been proposed to explain the effects of TTFields and other electrical therapies. Here, we present the first study of genome-wide expression of electrotherapy (delivered via TTFields or Deep Brain Stimulation (DBS)) on brain tumor cell lines. The effects of electric fields were assessed through gene expression arrays and combinational effects with chemotherapies. We observed that both DBS and TTFields significantly affected brain tumor cell line viability, with DBS promoting G0-phase accumulation and TTFields promoting G2-phase accumulation. Both treatments may be used to augment the efficacy of chemotherapy in vitro. Genome-wide expression assessment demonstrated significant overlap between the different electrical treatments, suggesting novel interactions with mitochondrial functioning and promoting endoplasmic reticulum stress. We demonstrate the in vitro efficacy of electric fields against adult and pediatric high-grade brain tumors and elucidate potential mechanisms of action for future study. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

20 pages, 6800 KiB  
Article
Functional In Vitro Assessment of VEGFA/NOTCH2 Signaling Pathway and pRB Proteasomal Degradation and the Clinical Relevance of Mucolipin TRPML2 Overexpression in Glioblastoma Patients
by Giorgio Santoni, Consuelo Amantini, Massimo Nabissi, Antonietta Arcella, Federica Maggi, Matteo Santoni and Maria Beatrice Morelli
Int. J. Mol. Sci. 2022, 23(2), 688; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23020688 - 08 Jan 2022
Cited by 4 | Viewed by 1650
Abstract
Glioblastoma (GBM) is the most malignant glioma with an extremely poor prognosis. It is characterized by high vascularization and its growth depends on the formation of new blood vessels. We have previously demonstrated that TRPML2 mucolipin channel expression increases with the glioma pathological [...] Read more.
Glioblastoma (GBM) is the most malignant glioma with an extremely poor prognosis. It is characterized by high vascularization and its growth depends on the formation of new blood vessels. We have previously demonstrated that TRPML2 mucolipin channel expression increases with the glioma pathological grade. Herein by ddPCR and Western blot we found that the silencing of TRPML2 inhibits expression of the VEGFA/Notch2 angiogenic pathway. Moreover, the VEGFA/Notch2 expression increased in T98 and U251 cells stimulated with the TRPML2 agonist, ML2-SA1, or by enforced-TRPML2 levels. In addition, changes in TRPML2 expression or ML2-SA1-induced stimulation, affected Notch2 activation and VEGFA release. An increased invasion capability, associated with a reduced VEGF/VEGFR2 expression and increased vimentin and CD44 epithelial-mesenchymal transition markers in siTRPML2, but not in enforced-TRPML2 or ML2-SA1-stimulated glioma cells, was demonstrated. Furthermore, an increased sensitivity to Doxorubicin cytotoxicity was demonstrated in siTRPML2, whereas ML2-SA1-treated GBM cells were more resistant. The role of proteasome in Cathepsin B-dependent and -independent pRB degradation in siTRPML2 compared with siGLO cells was studied. Finally, through Kaplan-Meier analysis, we found that high TRPML2 mRNA expression strongly correlates with short survival in GBM patients, supporting TRPML2 as a negative prognostic factor in GBM patients. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

15 pages, 1994 KiB  
Article
Sulforaphane Causes Cell Cycle Arrest and Apoptosis in Human Glioblastoma U87MG and U373MG Cell Lines under Hypoxic Conditions
by Giulia Sita, Agnese Graziosi, Patrizia Hrelia and Fabiana Morroni
Int. J. Mol. Sci. 2021, 22(20), 11201; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011201 - 18 Oct 2021
Cited by 3 | Viewed by 2312
Abstract
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor. The median survival rate from diagnosis ranges from 15 to 17 months because the tumor is resistant to most therapeutic strategies. GBM exhibits microvascular hyperplasia and pronounced necrosis triggered by hypoxia. [...] Read more.
Glioblastoma multiforme (GBM) is the most prevalent and aggressive primary brain tumor. The median survival rate from diagnosis ranges from 15 to 17 months because the tumor is resistant to most therapeutic strategies. GBM exhibits microvascular hyperplasia and pronounced necrosis triggered by hypoxia. Sulforaphane (SFN), an isothiocyanate derived from cruciferous vegetables, has already demonstrated the ability to inhibit cell proliferation, by provoking cell cycle arrest, and leading to apoptosis in many cell lines. In this study, we investigated the antineoplastic effects of SFN [20–80 μM for 48 h] in GBM cells under normoxic and hypoxic conditions. Cell viability assays, flow cytometry, and Western blot results revealed that SFN could induce apoptosis of GBM cells in a dose-dependent manner, under both conditions. In particular, SFN significantly induced caspase 3/7 activation and DNA fragmentation. Moreover, our results demonstrated that SFN suppressed GBM cells proliferation by arresting the cell cycle at the S-phase, also under hypoxic condition, and that these effects may be due in part to its ability to induce oxidative stress by reducing glutathione levels and to increase the phosphorylation of extracellular signal-regulated kinases (ERKs). Overall, we hypothesized that SFN treatment might serve as a potential therapeutic strategy, alone or in combination, against GBM. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

26 pages, 4263 KiB  
Article
Tumor-Associated Microglia/Macrophages as a Predictor for Survival in Glioblastoma and Temozolomide-Induced Changes in CXCR2 Signaling with New Resistance Overcoming Strategy by Combination Therapy
by Ruth M. Urbantat, Claudius Jelgersma, Susan Brandenburg, Melina Nieminen-Kelhä, Irina Kremenetskaia, Julia Zollfrank, Susanne Mueller, Kerstin Rubarth, Arend Koch, Peter Vajkoczy and Gueliz Acker
Int. J. Mol. Sci. 2021, 22(20), 11180; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011180 - 16 Oct 2021
Cited by 16 | Viewed by 2709
Abstract
Tumor recurrence is the main challenge in glioblastoma (GBM) treatment. Gold standard therapy temozolomide (TMZ) is known to induce upregulation of IL8/CXCL2/CXCR2 signaling that promotes tumor progression and angiogenesis. Our aim was to verify the alterations on this signaling pathway in human GBM [...] Read more.
Tumor recurrence is the main challenge in glioblastoma (GBM) treatment. Gold standard therapy temozolomide (TMZ) is known to induce upregulation of IL8/CXCL2/CXCR2 signaling that promotes tumor progression and angiogenesis. Our aim was to verify the alterations on this signaling pathway in human GBM recurrence and to investigate the impact of TMZ in particular. Furthermore, a combi-therapy of TMZ and CXCR2 antagonization was established to assess the efficacy and tolerability. First, we analyzed 76 matched primary and recurrent GBM samples with regard to various histological aspects with a focus on the role of TMZ treatment and the assessment of predictors of overall survival (OS). Second, the combi-therapy with TMZ and CXCR2-antagonization was evaluated in a syngeneic mouse tumor model with in-depth immunohistological investigations and subsequent gene expression analyses. We observed a significantly decreased infiltration of tumor-associated microglia/macrophages (TAM) in recurrent tumors, while a high TAM infiltration in primary tumors was associated with a reduced OS. Additionally, more patients expressed IL8 in recurrent tumors and TMZ therapy maintained CXCL2 expression. In mice, enhanced anti-tumoral effects were observed after combi-therapy. In conclusion, high TAM infiltration predicts a survival disadvantage, supporting findings of the tumor-promoting phenotype of TAMs. Furthermore, the combination therapy seemed to be promising to overcome CXCR2-mediated resistance. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

16 pages, 5846 KiB  
Article
Biobanked Glioblastoma Patient-Derived Organoids as a Precision Medicine Model to Study Inhibition of Invasion
by Emilie Darrigues, Edward H. Zhao, Annick De Loose, Madison P. Lee, Michael J. Borrelli, Robert L. Eoff, Deni S. Galileo, Narsimha R. Penthala, Peter A. Crooks and Analiz Rodriguez
Int. J. Mol. Sci. 2021, 22(19), 10720; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910720 - 03 Oct 2021
Cited by 12 | Viewed by 3116
Abstract
Glioblastoma (GBM) is highly resistant to treatment and invasion into the surrounding brain is a cancer hallmark that leads to recurrence despite surgical resection. With the emergence of precision medicine, patient-derived 3D systems are considered potentially robust GBM preclinical models. In this study, [...] Read more.
Glioblastoma (GBM) is highly resistant to treatment and invasion into the surrounding brain is a cancer hallmark that leads to recurrence despite surgical resection. With the emergence of precision medicine, patient-derived 3D systems are considered potentially robust GBM preclinical models. In this study, we screened a library of 22 anti-invasive compounds (i.e., NF-kB, GSK-3-B, COX-2, and tubulin inhibitors) using glioblastoma U-251 MG cell spheroids. We evaluated toxicity and invasion inhibition using a 3D Matrigel invasion assay. We next selected three compounds that inhibited invasion and screened them in patient-derived glioblastoma organoids (GBOs). We developed a platform using available macros for FIJI/ImageJ to quantify invasion from the outer margin of organoids. Our data demonstrated that a high-throughput invasion screening can be done using both an established cell line and patient-derived 3D model systems. Tubulin inhibitor compounds had the best efficacy with U-251 MG cells, however, in ex vivo patient organoids the results were highly variable. Our results indicate that the efficacy of compounds is highly related to patient intra and inter-tumor heterogeneity. These results indicate that such models can be used to evaluate personal oncology therapeutic strategies. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

16 pages, 4708 KiB  
Article
Construction of IL-13 Receptor α2-Targeting Resveratrol Nanoparticles against Glioblastoma Cells: Therapeutic Efficacy and Molecular Effects
by Xiao-Min Lin, Xiao-Xiao Shi, Le Xiong, Jun-Hua Nie, Hai-Shan Ye, Jin-Zi Du and Jia Liu
Int. J. Mol. Sci. 2021, 22(19), 10622; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910622 - 30 Sep 2021
Cited by 7 | Viewed by 1760
Abstract
Glioblastoma multiforme (GBM) is the most common lethal primary brain malignancy without reliable therapeutic drugs. IL-13Rα2 is frequently expressed in GBMs as a molecular marker. Resveratrol (Res) effectively inhibits GBM cell growth but has not been applied in vivo because of its low [...] Read more.
Glioblastoma multiforme (GBM) is the most common lethal primary brain malignancy without reliable therapeutic drugs. IL-13Rα2 is frequently expressed in GBMs as a molecular marker. Resveratrol (Res) effectively inhibits GBM cell growth but has not been applied in vivo because of its low brain bioavailability when administered systemically. A sustained-release and GBM-targeting resveratrol form may overcome this therapeutic dilemma. To achieve this goal, encapsulated Res 30 ± 4.8 nm IL-13Rα2-targeting nanoparticles (Pep-PP@Res) were constructed. Ultraviolet spectrophotometry revealed prolonged Res release (about 25%) from Pep-PP@Res in 48 h and fluorescent confocal microscopy showed the prolonged intracellular Res retention time of Pep-PP@Res (>24 h) in comparison with that of free Res (<4 h) and PP@Res (<4 h). MTT and EdU cell proliferation assays showed stronger suppressive effects of Pep-PP@Res on rat C6 GBM cells than that of PP@Res (p = 0.024) and Res (p = 0.009) when used twice for 4 h/day. Pep-PP@Res had little toxic effect on normal rat brain cells. The in vivo anti-glioblastoma effects of Res can be distinctly improved in the form of Pep-PP@Res nanoparticles via activating JNK signaling, upregulating proapoptosis gene expression and, finally, resulting in extensive apoptosis. Pep-PP@Res with sustained release and GBM-targeting properties would be suitable for in vivo management of GBMs. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

19 pages, 12507 KiB  
Article
Diosmin Inhibits Glioblastoma Growth through Inhibition of Autophagic Flux
by Yung-Lung Chang, Yao-Feng Li, Chung-Hsing Chou, Li-Chun Huang, Yi-Ping Wu, Ying Kao and Chia-Kuang Tsai
Int. J. Mol. Sci. 2021, 22(19), 10453; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms221910453 - 28 Sep 2021
Cited by 5 | Viewed by 2101
Abstract
Diosmin, a natural flavone glycoside acquired through dehydrogenation of the analogous flavanone glycoside hesperidin, is plentiful in many citrus fruits. Glioblastoma multiforme (GBM) is the most malignant primary brain tumor; the average survival time of GBM patients is less than 18 months after [...] Read more.
Diosmin, a natural flavone glycoside acquired through dehydrogenation of the analogous flavanone glycoside hesperidin, is plentiful in many citrus fruits. Glioblastoma multiforme (GBM) is the most malignant primary brain tumor; the average survival time of GBM patients is less than 18 months after standard treatment. The present study demonstrated that diosmin, which is able to cross the blood–brain barrier, inhibited GBM cell growth in vitro and in vivo. Diosmin also impeded migration and invasion by GBM8401and LN229 GBM cells by suppressing epithelial-mesenchymal transition, as indicated by increased expression of E-cadherin and decreased expression of Snail and Twist. Diosmin also suppressed autophagic flux, as indicated by increased expression of LC3-II and p62, and induced cell cycle arrest at G1 phase. Importantly, diosmin did not exert serious cytotoxic effects toward control SVG-p12 astrocytes, though it did reduce astrocyte viability at high concentrations. These findings provide potentially helpful support to the development of new therapies for the treatment of GBM. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

25 pages, 6798 KiB  
Article
Synchrotron-Based Fourier-Transform Infrared Micro-Spectroscopy (SR-FTIRM) Fingerprint of the Small Anionic Molecule Cobaltabis(dicarbollide) Uptake in Glioma Stem Cells
by Miquel Nuez-Martínez, Leire Pedrosa, Immaculada Martinez-Rovira, Ibraheem Yousef, Diouldé Diao, Francesc Teixidor, Elisabetta Stanzani, Fina Martínez-Soler, Avelina Tortosa, Àngels Sierra, José Juan Gonzalez and Clara Viñas
Int. J. Mol. Sci. 2021, 22(18), 9937; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22189937 - 14 Sep 2021
Cited by 9 | Viewed by 2973
Abstract
The anionic cobaltabis (dicarbollide) [3,3′-Co(1,2-C2B9H11)2], [o-COSAN], is the most studied icosahedral metallacarborane. The sodium salts of [o-COSAN] could be an ideal candidate for the anti-cancer treatment [...] Read more.
The anionic cobaltabis (dicarbollide) [3,3′-Co(1,2-C2B9H11)2], [o-COSAN], is the most studied icosahedral metallacarborane. The sodium salts of [o-COSAN] could be an ideal candidate for the anti-cancer treatment Boron Neutron Capture Therapy (BNCT) as it possesses the ability to readily cross biological membranes thereby producing cell cycle arrest in cancer cells. BNCT is a cancer therapy based on the potential of 10B atoms to produce α particles that cross tissues in which the 10B is accumulated without damaging the surrounding healthy tissues, after being irradiated with low energy thermal neutrons. Since Na[o-COSAN] displays a strong and characteristic ν(B-H) frequency in the infrared range 2.600–2.500 cm−1, we studied the uptake of Na[o-COSAN] followed by its interaction with biomolecules and its cellular biodistribution in two different glioma initiating cells (GICs), mesenchymal and proneural respectively, by using Synchrotron Radiation-Fourier Transform Infrared (FTIR) micro-spectroscopy (SR-FTIRM) facilities at the MIRAS Beamline of ALBA synchrotron light source. The spectroscopic data analysis from the bands in the regions of DNA, proteins, and lipids permitted to suggest that after its cellular uptake, Na[o-COSAN] strongly interacts with DNA strings, modifies proteins secondary structure and also leads to lipid saturation. The mapping suggests the nuclear localization of [o-COSAN], which according to reported Monte Carlo simulations may result in a more efficient cell-killing effect compared to that in a uniform distribution within the entire cell. In conclusion, we show pieces of evidence that at low doses, [o-COSAN] translocates GIC cells’ membranes and it alters the physiology of the cells, suggesting that Na[o-COSAN] is a promising agent to BNCT for glioblastoma cells. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

15 pages, 4871 KiB  
Article
In Vivo Study of the Efficacy and Safety of 5-Aminolevulinic Radiodynamic Therapy for Glioblastoma Fractionated Radiotherapy
by Junko Takahashi, Shinsuke Nagasawa, Motomichi Doi, Masamichi Takahashi, Yoshitaka Narita, Junkoh Yamamoto, Mitsushi J. Ikemoto and Hitoshi Iwahashi
Int. J. Mol. Sci. 2021, 22(18), 9762; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22189762 - 09 Sep 2021
Cited by 6 | Viewed by 2509
Abstract
To treat malignant glioma, standard fractionated radiotherapy (RT; 60 Gy/30 fractions over 6 weeks) was performed post-surgery in combination with temozolomide to improve overall survival. Malignant glioblastoma recurrence rate is extremely high, and most recurrent tumors originate from the excision cavity in the [...] Read more.
To treat malignant glioma, standard fractionated radiotherapy (RT; 60 Gy/30 fractions over 6 weeks) was performed post-surgery in combination with temozolomide to improve overall survival. Malignant glioblastoma recurrence rate is extremely high, and most recurrent tumors originate from the excision cavity in the high-dose irradiation region. In our previous study, protoporphyrin IX physicochemically enhanced reactive oxygen species generation by ionizing radiation and combined treatment with 5-aminolevulinic acid (5-ALA) and ionizing radiation, while radiodynamic therapy (RDT) improved tumor growth suppression in vivo in a melanoma mouse model. We examined the effect of 5-ALA RDT on the standard fractionated RT protocol using U251MG- or U87MG-bearing mice. 5-ALA was orally administered at 60 or 120 mg/kg, 4 h prior to irradiation. In both models, combined treatment with 5-ALA slowed tumor progression and promoted regression compared to treatment with ionizing radiation alone. The standard fractionated RT protocol of 60 Gy in 30 fractions with oral administration of 120 and 240 mg/kg 5-ALA, the human equivalent dose of photodynamic diagnosis, revealed no significant increase in toxicity to normal skin or brain tissue compared to ionizing radiation alone. Thus, RDT is expected to enhance RT treatment of glioblastoma without severe toxicity under clinically feasible conditions. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Graphical abstract

18 pages, 3439 KiB  
Article
Suppression of Proliferation of Human Glioblastoma Cells by Combined Phosphodiesterase and Multidrug Resistance-Associated Protein 1 Inhibition
by Liliya Kopanitsa, Maksym V. Kopanitsa, Dewi Safitri, Graham Ladds and David S. Bailey
Int. J. Mol. Sci. 2021, 22(18), 9665; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22189665 - 07 Sep 2021
Cited by 1 | Viewed by 1985
Abstract
The paucity of currently available therapies for glioblastoma multiforme requires novel approaches to the treatment of this brain tumour. Disrupting cyclic nucleotide-signalling through phosphodiesterase (PDE) inhibition may be a promising way of suppressing glioblastoma growth. Here, we examined the effects of 28 PDE [...] Read more.
The paucity of currently available therapies for glioblastoma multiforme requires novel approaches to the treatment of this brain tumour. Disrupting cyclic nucleotide-signalling through phosphodiesterase (PDE) inhibition may be a promising way of suppressing glioblastoma growth. Here, we examined the effects of 28 PDE inhibitors, covering all the major PDE classes, on the proliferation of the human U87MG, A172 and T98G glioblastoma cells. The PDE10A inhibitors PF-2545920, PQ10 and papaverine, the PDE3/4 inhibitor trequinsin and the putative PDE5 inhibitor MY-5445 potently decreased glioblastoma cell proliferation. The synergistic suppression of glioblastoma cell proliferation was achieved by combining PF-2545920 and MY-5445. Furthermore, a co-incubation with drugs that block the activity of the multidrug resistance-associated protein 1 (MRP1) augmented these effects. In particular, a combination comprising the MRP1 inhibitor reversan, PF-2545920 and MY-5445, all at low micromolar concentrations, afforded nearly complete inhibition of glioblastoma cell growth. Thus, the potent suppression of glioblastoma cell viability may be achieved by combining MRP1 inhibitors with PDE inhibitors at a lower toxicity than that of the standard chemotherapeutic agents, thereby providing a new combination therapy for this challenging malignancy. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

25 pages, 8271 KiB  
Article
Behind Brain Metastases Formation: Cellular and Molecular Alterations and Blood–Brain Barrier Disruption
by Joana Godinho-Pereira, Ana Rita Garcia, Inês Figueira, Rui Malhó and Maria Alexandra Brito
Int. J. Mol. Sci. 2021, 22(13), 7057; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22137057 - 30 Jun 2021
Cited by 15 | Viewed by 3182
Abstract
Breast cancer (BC) brain metastases is a life-threatening condition to which accounts the poor understanding of BC cells’ (BCCs) extravasation into the brain, precluding the development of preventive strategies. Thus, we aimed to unravel the players involved in the interaction between BCCs and [...] Read more.
Breast cancer (BC) brain metastases is a life-threatening condition to which accounts the poor understanding of BC cells’ (BCCs) extravasation into the brain, precluding the development of preventive strategies. Thus, we aimed to unravel the players involved in the interaction between BCCs and blood–brain barrier (BBB) endothelial cells underlying BBB alterations and the transendothelial migration of malignant cells. We used brain microvascular endothelial cells (BMECs) as a BBB in vitro model, under conditions mimicking shear stress to improve in vivo-like BBB features. Mixed cultures were performed by the addition of fluorescently labelled BCCs to distinguish individual cell populations. BCC–BMEC interaction compromised BBB integrity, as revealed by junctional proteins (β-catenin and zonula occludens-1) disruption and caveolae (caveolin-1) increase, reflecting paracellular and transcellular hyperpermeability, respectively. Both BMECs and BCCs presented alterations in the expression pattern of connexin 43, suggesting the involvement of the gap junction protein. Myosin light chain kinase and phosphorylated myosin light chain were upregulated, revealing the involvement of the endothelial cytoskeleton in the extravasation process. β4-Integrin and focal adhesion kinase were colocalised in malignant cells, reflecting molecular interaction. Moreover, BCCs exhibited invadopodia, attesting migratory properties. Collectively, hub players involved in BC brain metastases formation were unveiled, disclosing possible therapeutic targets for metastases prevention. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

18 pages, 5461 KiB  
Article
NBM-BMX, an HDAC8 Inhibitor, Overcomes Temozolomide Resistance in Glioblastoma Multiforme by Downregulating the β-Catenin/c-Myc/SOX2 Pathway and Upregulating p53-Mediated MGMT Inhibition
by Cheng-Yu Tsai, Huey-Jiun Ko, Shean-Jaw Chiou, Yu-Ling Lai, Chia-Chung Hou, Tehseen Javaria, Zi-Yi Huang, Tai-Shan Cheng, Tsung-I Hsu, Jian-Ying Chuang, Aij-Lie Kwan, Tsung-Hsien Chuang, Chi-Ying F. Huang, Joon-Khim Loh and Yi-Ren Hong
Int. J. Mol. Sci. 2021, 22(11), 5907; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22115907 - 31 May 2021
Cited by 12 | Viewed by 3394
Abstract
Although histone deacetylase 8 (HDAC8) plays a role in glioblastoma multiforme (GBM), whether its inhibition facilitates the treatment of temozolomide (TMZ)-resistant GBM (GBM-R) remains unclear. By assessing the gene expression profiles from short hairpin RNA of HDAC8 in the new version of Connectivity [...] Read more.
Although histone deacetylase 8 (HDAC8) plays a role in glioblastoma multiforme (GBM), whether its inhibition facilitates the treatment of temozolomide (TMZ)-resistant GBM (GBM-R) remains unclear. By assessing the gene expression profiles from short hairpin RNA of HDAC8 in the new version of Connectivity Map (CLUE) and cells treated by NBM-BMX (BMX)-, an HDAC8 inhibitor, data analysis reveals that the Wnt signaling pathway and apoptosis might be the underlying mechanisms in BMX-elicited treatment. This study evaluated the efficacy of cotreatment with BMX and TMZ in GBM-R cells. We observed that cotreatment with BMX and TMZ could overcome resistance in GBM-R cells and inhibit cell viability, markedly inhibit cell proliferation, and then induce cell cycle arrest and apoptosis. In addition, the expression level of β-catenin was reversed by proteasome inhibitor via the β-catenin/ GSK3β signaling pathway to reduce the expression level of c-Myc and cyclin D1 in GBM-R cells. BMX and TMZ cotreatment also upregulated WT-p53 mediated MGMT inhibition, thereby triggering the activation of caspase-3 and eventually leading to apoptosis in GBM-R cells. Moreover, BMX and TMZ attenuated the expression of CD133, CD44, and SOX2 in GBM-R cells. In conclusion, BMX overcomes TMZ resistance by enhancing TMZ-mediated cytotoxic effect by downregulating the β-catenin/c-Myc/SOX2 signaling pathway and upregulating WT-p53 mediated MGMT inhibition. These findings indicate a promising drug combination for precision personal treating of TMZ-resistant WT-p53 GBM cells. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

14 pages, 2988 KiB  
Article
Combining Magnetic Resonance Imaging with Systemic Monocyte Evaluation for the Implementation of GBM Management
by Carolina Giordano, Giovanni Sabatino, Simona Romano, Giuseppe Maria Della Pepa, Martina Tufano, Quintino Giorgio D’Alessandris, Simone Cottonaro, Marco Gessi, Mario Balducci, Maria Fiammetta Romano, Alessandro Olivi, Simona Gaudino and Cesare Colosimo
Int. J. Mol. Sci. 2021, 22(7), 3797; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073797 - 06 Apr 2021
Cited by 6 | Viewed by 2447
Abstract
Magnetic resonance imaging (MRI) is the gold standard for glioblastoma (GBM) patient evaluation. Additional non-invasive diagnostic modalities are needed. GBM is heavily infiltrated with tumor-associated macrophages (TAMs) that can be found in peripheral blood. FKBP51s supports alternative-macrophage polarization. Herein, we assessed FKBP51s expression [...] Read more.
Magnetic resonance imaging (MRI) is the gold standard for glioblastoma (GBM) patient evaluation. Additional non-invasive diagnostic modalities are needed. GBM is heavily infiltrated with tumor-associated macrophages (TAMs) that can be found in peripheral blood. FKBP51s supports alternative-macrophage polarization. Herein, we assessed FKBP51s expression in circulating monocytes from 14 GBM patients. The M2 monocyte phenotype was investigated by qPCR and flow cytometry using antibodies against PD-L1, CD163, FKBP51s, and CD14. MRI assessed morphologic features of the tumors that were aligned to flow cytometry data. PD-L1 expression on circulating monocytes correlated with MRI tumor necrosis score. A wider expansion in circulating CD163/monocytes was measured. These monocytes resulted in a dramatic decrease in patients with an MRI diagnosis of complete but not partial surgical removal of the tumor. Importantly, in patients with residual tumor, most of the peripheral monocytes that in the preoperative stage were CD163/FKBP51s− had turned into CD163/FKBP51s+. After Stupp therapy, CD163/FKBP51s+ monocytes were almost absent in a case of pseudoprogression, while two patients with stable or true disease progression showed sustained levels in such circulating monocytes. Our work provides preliminary but meaningful and novel results that deserve to be confirmed in a larger patient cohort, in support of potential usefulness in GBM monitoring of CD163/FKBP51s/CD14 immunophenotype in adjunct to MRI. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

18 pages, 2296 KiB  
Article
Successful Partnerships: Exploring the Potential of Immunogenic Signals Triggered by TMZ, CX-4945, and Combined Treatment in GL261 Glioblastoma Cells
by Lucía Villamañan, Laura Martínez-Escardó, Carles Arús, Victor J. Yuste and Ana P. Candiota
Int. J. Mol. Sci. 2021, 22(7), 3453; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22073453 - 26 Mar 2021
Cited by 7 | Viewed by 2480
Abstract
Background: The relevance of the cancer immune cycle in therapy response implies that successful treatment may trigger the exposure or the release of immunogenic signals. Previous results with the preclinical GL261 glioblastoma (GB) showed that combination treatment of temozolomide (TMZ) + CX-4945 (protein [...] Read more.
Background: The relevance of the cancer immune cycle in therapy response implies that successful treatment may trigger the exposure or the release of immunogenic signals. Previous results with the preclinical GL261 glioblastoma (GB) showed that combination treatment of temozolomide (TMZ) + CX-4945 (protein kinase CK2 inhibitor) outperformed single treatments, provided an immune-friendly schedule was followed. Our purpose was to study possible immunogenic signals released in vitro by GB cells. Methods: GL261 GB cells were treated with TMZ and CX-4945 at different concentrations (25 µM–4 mM) and time frames (12–72 h). Cell viability was measured with Trypan Blue and propidium iodide. Calreticulin exposure was assessed with immunofluorescence, and ATP release was measured with bioluminescence. Results: TMZ showed cytostatic rather than cytotoxic effects, while CX-4945 showed remarkable cytotoxic effects already at low concentrations. Calreticulin exposure after 24 h was detected with TMZ treatment, as well as TMZ/CX-4945 low concentration combined treatment. ATP release was significantly higher with CX-4945, especially at high concentrations, as well as with TMZ/CX-4945. Conclusions: combined treatment may produce the simultaneous release of two potent immunogenic signals, which can explain the outperformance over single treatments in vivo. A word of caution may be raised since in vitro conditions are not able to mimic pharmacokinetics observed in vivo fully. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Graphical abstract

Review

Jump to: Research

34 pages, 4454 KiB  
Review
Therapeutic Options in Neuro-Oncology
by Mariana Afonso and Maria Alexandra Brito
Int. J. Mol. Sci. 2022, 23(10), 5351; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms23105351 - 11 May 2022
Cited by 9 | Viewed by 3700
Abstract
One of the biggest challenges in neuro-oncology is understanding the complexity of central nervous system tumors, such as gliomas, in order to develop suitable therapeutics. Conventional therapies in malignant gliomas reconcile surgery and radiotherapy with the use of chemotherapeutic options such as temozolomide, [...] Read more.
One of the biggest challenges in neuro-oncology is understanding the complexity of central nervous system tumors, such as gliomas, in order to develop suitable therapeutics. Conventional therapies in malignant gliomas reconcile surgery and radiotherapy with the use of chemotherapeutic options such as temozolomide, chloroethyl nitrosoureas and the combination therapy of procarbazine, lomustine and vincristine. With the unraveling of deregulated cancer cell signaling pathways, targeted therapies have been developed. The most affected signaling pathways in glioma cells involve tyrosine kinase receptors and their downstream pathways, such as the phosphatidylinositol 3-kinases (PI3K/AKT/mTOR) and mitogen-activated protein kinase pathways (MAPK). MAPK pathway inhibitors include farnesyl transferase inhibitors, Ras kinase inhibitors and mitogen-activated protein extracellular regulated kinase (MEK) inhibitors, while PI3K/AKT/mTOR pathway inhibitors are divided into pan-inhibitors, PI3K/mTOR dual inhibitors and AKT inhibitors. The relevance of the immune system in carcinogenesis has led to the development of immunotherapy, through vaccination, blocking of immune checkpoints, oncolytic viruses, and adoptive immunotherapy using chimeric antigen receptor T cells. In this article we provide a comprehensive review of the signaling pathways underlying malignant transformation, the therapies currently used in the treatment of malignant gliomas and further explore therapies under development, including several ongoing clinical trials. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

16 pages, 687 KiB  
Review
Genetically Modified Cellular Therapies for Malignant Gliomas
by Michael Kilian, Theresa Bunse, Wolfgang Wick, Michael Platten and Lukas Bunse
Int. J. Mol. Sci. 2021, 22(23), 12810; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222312810 - 26 Nov 2021
Cited by 9 | Viewed by 3176
Abstract
Despite extensive preclinical research on immunotherapeutic approaches, malignant glioma remains a devastating disease of the central nervous system for which standard of care treatment is still confined to resection and radiochemotherapy. For peripheral solid tumors, immune checkpoint inhibition has shown substantial clinical benefit, [...] Read more.
Despite extensive preclinical research on immunotherapeutic approaches, malignant glioma remains a devastating disease of the central nervous system for which standard of care treatment is still confined to resection and radiochemotherapy. For peripheral solid tumors, immune checkpoint inhibition has shown substantial clinical benefit, while promising preclinical results have yet failed to translate into clinical efficacy for brain tumor patients. With the advent of high-throughput sequencing technologies, tumor antigens and corresponding T cell receptors (TCR) and antibodies have been identified, leading to the development of chimeric antigen receptors (CAR), which are comprised of an extracellular antibody part and an intracellular T cell receptor signaling part, to genetically engineer T cells for antigen recognition. Due to efficacy in other tumor entities, a plethora of CARs has been designed and tested for glioma, with promising signs of biological activity. In this review, we describe glioma antigens that have been targeted using CAR T cells preclinically and clinically, review their drawbacks and benefits, and illustrate how the emerging field of transgenic TCR therapy can be used as a potent alternative for cell therapy of glioma overcoming antigenic limitations. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

15 pages, 1972 KiB  
Review
Zika Virus: A New Therapeutic Candidate for Glioblastoma Treatment
by Maria Giovanna Francipane, Bruno Douradinha, Cinzia Maria Chinnici, Giovanna Russelli, Pier Giulio Conaldi and Gioacchin Iannolo
Int. J. Mol. Sci. 2021, 22(20), 10996; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222010996 - 12 Oct 2021
Cited by 13 | Viewed by 2950
Abstract
Glioblastoma (GBM) is the most aggressive among the neurological tumors. At present, no chemotherapy or radiotherapy regimen is associated with a positive long-term outcome. In the majority of cases, the tumor recurs within 32–36 weeks of initial treatment. The recent discovery that Zika [...] Read more.
Glioblastoma (GBM) is the most aggressive among the neurological tumors. At present, no chemotherapy or radiotherapy regimen is associated with a positive long-term outcome. In the majority of cases, the tumor recurs within 32–36 weeks of initial treatment. The recent discovery that Zika virus (ZIKV) has an oncolytic action against GBM has brought hope for the development of new therapeutic approaches. ZIKV is an arbovirus of the Flaviviridae family, and its infection during development has been associated with central nervous system (CNS) malformations, including microcephaly, through the targeting of neural stem/progenitor cells (NSCs/NPCs). This finding has led various groups to evaluate ZIKV’s effects against glioblastoma stem cells (GSCs), supposedly responsible for GBM onset, progression, and therapy resistance. While preliminary data support ZIKV tropism toward GSCs, a more accurate study of ZIKV mechanisms of action is fundamental in order to launch ZIKV-based clinical trials for GBM patients. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

22 pages, 1593 KiB  
Review
Radiotherapy of High-Grade Gliomas: First Half of 2021 Update with Special Reference to Radiosensitization Studies
by Guido Frosina
Int. J. Mol. Sci. 2021, 22(16), 8942; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22168942 - 19 Aug 2021
Cited by 10 | Viewed by 3516
Abstract
Albeit the effort to develop targeted therapies for patients with high-grade gliomas (WHO grades III and IV) is evidenced by hundreds of current clinical trials, radiation remains one of the few effective therapeutic options for them. This review article analyzes the updates on [...] Read more.
Albeit the effort to develop targeted therapies for patients with high-grade gliomas (WHO grades III and IV) is evidenced by hundreds of current clinical trials, radiation remains one of the few effective therapeutic options for them. This review article analyzes the updates on the topic “radiotherapy of high-grade gliomas” during the period 1 January 2021–30 June 2021. The high number of articles retrieved in PubMed using the search terms (“gliom* and radio*”) and manually selected for relevance indicates the feverish research currently ongoing on the subject. During the last semester, significant advances were provided in both the preclinical and clinical settings concerning the diagnosis and prognosis of high-grade gliomas, their radioresistance, and the inevitable side effects of their treatment with radiation. The novel information concerning tumor radiosensitization was of special interest in terms of therapeutic perspective and was discussed in detail. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

16 pages, 712 KiB  
Review
The Association of Human Herpesviruses with Malignant Brain Tumor Pathology and Therapy: Two Sides of a Coin
by Evita Athanasiou, Antonios N. Gargalionis, Fotini Boufidou and Athanassios Tsakris
Int. J. Mol. Sci. 2021, 22(5), 2250; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22052250 - 24 Feb 2021
Cited by 8 | Viewed by 2692
Abstract
The role of certain viruses in malignant brain tumor development remains controversial. Experimental data demonstrate that human herpesviruses (HHVs), particularly cytomegalovirus (CMV), Epstein–Barr virus (EBV) and human herpes virus 6 (HHV-6), are implicated in brain tumor pathology, although their direct role has not [...] Read more.
The role of certain viruses in malignant brain tumor development remains controversial. Experimental data demonstrate that human herpesviruses (HHVs), particularly cytomegalovirus (CMV), Epstein–Barr virus (EBV) and human herpes virus 6 (HHV-6), are implicated in brain tumor pathology, although their direct role has not yet been proven. CMV is present in most gliomas and medulloblastomas and is known to facilitate oncomodulation and/or immunomodulation, thus promoting cancer cell proliferation, invasion, apoptosis, angiogenesis, and immunosuppression. EBV and HHV-6 have also been detected in brain tumors and high-grade gliomas, showing high rates of expression and an inflammatory potential. On the other hand, due to the neurotropic nature of HHVs, novel studies have highlighted the engagement of such viruses in the development of new immunotherapeutic approaches in the context of oncolytic viral treatment and vaccine-based strategies against brain tumors. This review provides a comprehensive evaluation of recent scientific data concerning the emerging dual role of HHVs in malignant brain pathology, either as potential causative agents or as immunotherapeutic tools in the fight against these devastating diseases. Full article
(This article belongs to the Special Issue Frontiers in Neuro-Oncology)
Show Figures

Figure 1

Back to TopTop