1
|
Zhang Y, Chen B, Liu R, Mei W, Lin Y. Deciphering glioblastoma pathogenesis: Insights from mitophagy dysregulation and SNX7 as a therapeutic target. Brain Res Bull 2025; 220:111184. [PMID: 39736337 DOI: 10.1016/j.brainresbull.2024.111184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/25/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND Glioblastoma is a highly aggressive and invasive brain tumor with an extremely poor prognosis. The aims of the present study are to investigate the pathogenesis of glioblastoma and identify potential therapeutic targets. METHODS We performed a systematic analysis of gene expression data from multiple datasets, including GEO and TCGA, to identify hub genes and pathways associated with glioblastoma progression. Bioinformatics tools were utilized to analyze differential gene expression, pathway enrichment and survival prognosis. Both in vitro and in vivo functional experiments were conducted to validate biological roles of SNX7. RESULTS Pathway analysis revealed significant enrichment of the mitophagy pathway in glioblastoma, indicating its critical role in tumor development. We identified 12 hub genes associated with glioblastoma prognosis, with high-risk patients having worse survival outcomes. Among the hub gene set, sorting nexin 7 (SNX7) was found to be the most significant regulator of glioblastoma progression. Our results also demonstrated that SNX7 expression is associated with tumor ferroptosis and genomic variations, representing potential biomarkers for clinical diagnosis and treatment. Furthermore, functional experiments confirmed that SNX7 promotes glioblastoma cell proliferation, invasion and survival by inhibiting protective mitophagy. CONCLUSION Our results highlight the importance of mitophagy dysregulation in the pathogenesis of glioblastoma and identify SNX7 as a novel therapeutic target. Further research is needed to elucidate the underlying mechanisms of SNX7 in glioblastoma and validate its clinical significance. These findings may facilitate the development of personalized treatment strategies and improve outcomes for glioblastoma patients.
Collapse
Affiliation(s)
- Yuanlong Zhang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China
| | - Binghong Chen
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China
| | - Renfu Liu
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China
| | - Wenzhong Mei
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China.
| | - Yuanxiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China; Fujian Provincial Institutes of Brain Disorders and Brain Sciences, First Affiliated Hospital of Fujian Medical University, Fujian, Fuzhou 350005, China.
| |
Collapse
|
2
|
Wu Q, Berglund AE, Macaulay RJ, Etame AB. The Role of Mesenchymal Reprogramming in Malignant Clonal Evolution and Intra-Tumoral Heterogeneity in Glioblastoma. Cells 2024; 13:942. [PMID: 38891074 PMCID: PMC11171993 DOI: 10.3390/cells13110942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma (GBM) is the most common yet uniformly fatal adult brain cancer. Intra-tumoral molecular and cellular heterogeneities are major contributory factors to therapeutic refractoriness and futility in GBM. Molecular heterogeneity is represented through molecular subtype clusters whereby the proneural (PN) subtype is associated with significantly increased long-term survival compared to the highly resistant mesenchymal (MES) subtype. Furthermore, it is universally recognized that a small subset of GBM cells known as GBM stem cells (GSCs) serve as reservoirs for tumor recurrence and progression. The clonal evolution of GSC molecular subtypes in response to therapy drives intra-tumoral heterogeneity and remains a critical determinant of GBM outcomes. In particular, the intra-tumoral MES reprogramming of GSCs using current GBM therapies has emerged as a leading hypothesis for therapeutic refractoriness. Preventing the intra-tumoral divergent evolution of GBM toward the MES subtype via new treatments would dramatically improve long-term survival for GBM patients and have a significant impact on GBM outcomes. In this review, we examine the challenges of the role of MES reprogramming in the malignant clonal evolution of glioblastoma and provide future perspectives for addressing the unmet therapeutic need to overcome resistance in GBM.
Collapse
Affiliation(s)
- Qiong Wu
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Robert J. Macaulay
- Departments of Anatomic Pathology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Arnold B. Etame
- Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
3
|
Zhao X, Ni S, Song Y, Hu K. Intranasal delivery of Borneol/R8dGR peptide modified PLGA nanoparticles co-loaded with curcumin and cisplatin alleviate hypoxia in pediatric brainstem glioma which improves the synergistic therapy. J Control Release 2023; 362:121-137. [PMID: 37633362 DOI: 10.1016/j.jconrel.2023.08.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/20/2023] [Accepted: 08/24/2023] [Indexed: 08/28/2023]
Abstract
Cisplatin (cis) is a first-line chemotherapeutic used for the treatment of intractable pediatric brainstem glioma (PBSG). Its therapeutic effect in PBSG is, however, critically challenged by the hypoxic microenvironment of the tumor and the presence of the blood brain barrier (BBB). Herein, we report on the intranasal administration of borneol (Bo)/R8dGR peptide modified PLGA based nanoparticles (NP) co-loaded with curcumin and cisplatin (cur/cis). We observed that borneol modification improved the brain penetration of the nanoparticles by reduction of the expression of ZO-1 and occludin in nasal mucosa, while the R8dGR peptide modification allowed the targeting of the NP through the binding on integrin αvβ3 receptors which are present on PBSG cells. Following intranasal administration, BoR-cur/cis-NP attenuated hypoxia in the PBSG microenvironment and reduced angiogenesis, which prolonged survival of GL261-bearing PBSG mice. Therefore, intranasal administration of BoR-cur/cis-NP, which deeply penetrate PBSG, is an encouraging strategy to attenuate hypoxia which potentiates the efficacy of cisplatin in the treatment of PBSG.
Collapse
Affiliation(s)
- Xiao Zhao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Shuting Ni
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Yangjie Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, PR China.
| |
Collapse
|
4
|
Schnöller LE, Piehlmaier D, Weber P, Brix N, Fleischmann DF, Nieto AE, Selmansberger M, Heider T, Hess J, Niyazi M, Belka C, Lauber K, Unger K, Orth M. Systematic in vitro analysis of therapy resistance in glioblastoma cell lines by integration of clonogenic survival data with multi-level molecular data. Radiat Oncol 2023; 18:51. [PMID: 36906590 PMCID: PMC10007763 DOI: 10.1186/s13014-023-02241-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 03/13/2023] Open
Abstract
Despite intensive basic scientific, translational, and clinical efforts in the last decades, glioblastoma remains a devastating disease with a highly dismal prognosis. Apart from the implementation of temozolomide into the clinical routine, novel treatment approaches have largely failed, emphasizing the need for systematic examination of glioblastoma therapy resistance in order to identify major drivers and thus, potential vulnerabilities for therapeutic intervention. Recently, we provided proof-of-concept for the systematic identification of combined modality radiochemotherapy treatment vulnerabilities via integration of clonogenic survival data upon radio(chemo)therapy with low-density transcriptomic profiling data in a panel of established human glioblastoma cell lines. Here, we expand this approach to multiple molecular levels, including genomic copy number, spectral karyotyping, DNA methylation, and transcriptome data. Correlation of transcriptome data with inherent therapy resistance on the single gene level yielded several candidates that were so far underappreciated in this context and for which clinically approved drugs are readily available, such as the androgen receptor (AR). Gene set enrichment analyses confirmed these results, and identified additional gene sets, including reactive oxygen species detoxification, mammalian target of rapamycin complex 1 (MTORC1) signaling, and ferroptosis/autophagy-related regulatory circuits to be associated with inherent therapy resistance in glioblastoma cells. To identify pharmacologically accessible genes within those gene sets, leading edge analyses were performed yielding candidates with functions in thioredoxin/peroxiredoxin metabolism, glutathione synthesis, chaperoning of proteins, prolyl hydroxylation, proteasome function, and DNA synthesis/repair. Our study thus confirms previously nominated targets for mechanism-based multi-modal glioblastoma therapy, provides proof-of-concept for this workflow of multi-level data integration, and identifies novel candidates for which pharmacological inhibitors are readily available and whose targeting in combination with radio(chemo)therapy deserves further examination. In addition, our study also reveals that the presented workflow requires mRNA expression data, rather than genomic copy number or DNA methylation data, since no stringent correlation between these data levels could be observed. Finally, the data sets generated in the present study, including functional and multi-level molecular data of commonly used glioblastoma cell lines, represent a valuable toolbox for other researchers in the field of glioblastoma therapy resistance.
Collapse
Affiliation(s)
- Leon Emanuel Schnöller
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Daniel Piehlmaier
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Peter Weber
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Nikko Brix
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Daniel Felix Fleischmann
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Edward Nieto
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany
| | - Martin Selmansberger
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Theresa Heider
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany
| | - Julia Hess
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany
| | - Maximilian Niyazi
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Bavarian Cancer Research Center (BKFZ), Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium (DKTK), Munich, Germany.,Bavarian Cancer Research Center (BKFZ), Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.,German Cancer Consortium (DKTK), Munich, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics (ZYTO), Helmholtz Center Munich, German Research Center for Environmental Health GmbH, 85764, Neuherberg, Germany. .,Clinical Cooperation Group 'Personalized Radiotherapy in Head and Neck Cancer' Helmholtz Center Munich, German Research Center for Environmental Health GmbH, Neuherberg, Germany.
| | - Michael Orth
- Department of Radiation Oncology, University Hospital, LMU München, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
5
|
Bhaduri S, Kelly CL, Lesbats C, Sharkey J, Ressel L, Mukherjee S, Platt MD, Delikatny EJ, Poptani H. Metabolic changes in glioblastomas in response to choline kinase inhibition: In vivo MRS in rodent models. NMR IN BIOMEDICINE 2023; 36:e4855. [PMID: 36269130 PMCID: PMC10078495 DOI: 10.1002/nbm.4855] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 06/16/2023]
Abstract
Changes in glioblastoma (GBM) metabolism was investigated in response to JAS239, a choline kinase inhibitor, using MRS. In addition to the inhibition of phosphocholine synthesis, we investigated changes in other key metabolic pathways associated with GBM progression and treatment response. Three syngeneic rodent models of GBM were used: F98 (N = 12) and 9L (N = 8) models in rats and GL261 (N = 10) in mice. Rodents were intracranially injected with GBM cells in the right cortex and tumor growth was monitored using T2 -weighted images. Animals were treated once daily with intraperitoneal injections of 4 mg/kg JAS239 (F98 rats, n = 6; 9L rats, n = 6; GL261 mice, n = 5) or saline (control group, F98 rats, n = 6; 9L rats, n = 2; GL261 mice, n = 5) for five consecutive days. Single voxel spectra were acquired on Days 0 (T0, baseline) and 6 (T6, end of treatment) from the tumor as well as the contralateral normal brain using a PRESS sequence. Changes in metabolite ratios (tCho/tCr, tCho/NAA, mI/tCr, Glx/tCr and (Lip + Lac)/Cr) were used to assess metabolic pathway alterations in response to JAS239. Tumor growth arrest was noted in all models in response to JAS239 treatment compared with saline-treated animals, with a significant reduction (p < 0.05) in the F98 model. A reduction in tCho/tCr was observed with JAS239 treatment in all GBM models, indicating reduced phospholipid metabolism, with the highest reduction in 9L followed by GL261 and F98 tumors. A significant reduction (p < 0.05) in the tCho/NAA ratio was observed in the 9L model. A significant reduction in mI/tCr (p < 0.05) was found in JAS239-treated F98 tumors compared with the saline-treated animals. A non-significant trend of reduction in Glx/tCr was observed only in F98 and 9L tumors. JAS239-treated F98 tumors also showed a significant increase in Lip + Lac (p < 0.05), indicating increased cell death. This study demonstrated the utility of MRS in assessing metabolic changes in GBM in response to choline kinase inhibition.
Collapse
Affiliation(s)
- Sourav Bhaduri
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Claire Louise Kelly
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Clémentine Lesbats
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
- Division of Radiotherapy and ImagingThe Institute of Cancer ResearchLondonUK
| | - Jack Sharkey
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Lorenzo Ressel
- Department of Veterinary Anatomy Physiology and PathologyUniversity of LiverpoolChesterUK
| | - Soham Mukherjee
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Mark David Platt
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| | - Edward J. Delikatny
- Department of Radiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Harish Poptani
- Centre for Preclinical Imaging, Department of Molecular and Clinical Cancer MedicineUniversity of LiverpoolLiverpoolUK
| |
Collapse
|
6
|
Gubanova NV, Orlova NG, Dergilev AI, Oparina NY, Orlov YL. Glioblastoma gene network reconstruction and ontology analysis by online bioinformatics tools. J Integr Bioinform 2021; 18:jib-2021-0031. [PMID: 34783229 PMCID: PMC8709738 DOI: 10.1515/jib-2021-0031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most aggressive type of brain tumors resistant to a number of antitumor drugs. The problem of therapy and drug treatment course is complicated by extremely high heterogeneity in the benign cell populations, the random arrangement of tumor cells, and polymorphism of their nuclei. The pathogenesis of gliomas needs to be studied using modern cellular technologies, genome- and transcriptome-wide technologies of high-throughput sequencing, analysis of gene expression on microarrays, and methods of modern bioinformatics to find new therapy targets. Functional annotation of genes related to the disease could be retrieved based on genetic databases and cross-validated by integrating complementary experimental data. Gene network reconstruction for a set of genes (proteins) proved to be effective approach to study mechanisms underlying disease progression. We used online bioinformatics tools for annotation of gene list for glioma, reconstruction of gene network and comparative analysis of gene ontology categories. The available tools and the databases for glioblastoma gene analysis are discussed together with the recent progress in this field.
Collapse
Affiliation(s)
- Natalya V Gubanova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nina G Orlova
- Financial University under the Government of the Russian Federation, 119991 Moscow, Russia.,Moscow State Technical University of Civil Aviation, 125993 Moscow, Russia
| | | | | | - Yuriy L Orlov
- Novosibirsk State University, 630090 Novosibirsk, Russia.,The Digital Health Institute, I.M.Sechenov First Moscow State Medical University of the Russian Ministry of Health, 119991 Moscow, Russia
| |
Collapse
|
7
|
Degorre C, Tofilon P, Camphausen K, Mathen P. Bench to bedside radiosensitizer development strategy for newly diagnosed glioblastoma. Radiat Oncol 2021; 16:191. [PMID: 34583727 PMCID: PMC8480070 DOI: 10.1186/s13014-021-01918-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/15/2021] [Indexed: 11/10/2022] Open
Abstract
Glioblastoma is the most common primary brain malignancy and carries with it a poor prognosis. New agents are urgently needed, however nearly all Phase III trials of GBM patients of the past 25 years have failed to demonstrate improvement in outcomes. In 2019, the National Cancer Institute Clinical Trials and Translational Research Advisory Committee (CTAC) Glioblastoma Working Group (GBM WG) identified 5 broad areas of research thought to be important in the development of new herapeutics for GBM. Among those was optimizing radioresponse for GBM in situ. One such strategy to increase radiation efficacy is the addition of a radiosensitizer to improve the therapeutic ratio by enhancing tumor sensitivity while ideally having minimal to no effect on normal tissue. Historically the majority of trials using radiosensitizers have been unsuccessful, but they provide important guidance in what is required to develop agents more efficiently. Improved target selection is essential for a drug to provide maximal benefit, and once that target is identified it must be validated through pre-clinical studies. Careful selection of appropriate in vitro and in vivo models to demonstrate increased radiosensitivity and suitable bioavailability are then necessary to prove that a drug warrants advancement to clinical investigation. Once investigational agents are validated pre-clinically, patient trials require consistency both in terms of planning study design as well as reporting efficacy and toxicity in order to assess the potential benefit of the drug. Through this paper we hope to outline strategies for developing effective radiosensitizers against GBM using as models the examples of XPO1 inhibitors and HDAC inhibitors developed from our own lab.
Collapse
Affiliation(s)
- Charlotte Degorre
- Radiation Oncology Branch, National Cancer Institute, Bldg. 10, Rm B2-3500, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Philip Tofilon
- Radiation Oncology Branch, National Cancer Institute, Bldg. 10, Rm B2-3500, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bldg. 10, Rm B2-3500, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Peter Mathen
- Radiation Oncology Branch, National Cancer Institute, Bldg. 10, Rm B2-3500, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| |
Collapse
|
8
|
Comba A, Faisal SM, Varela ML, Hollon T, Al-Holou WN, Umemura Y, Nunez FJ, Motsch S, Castro MG, Lowenstein PR. Uncovering Spatiotemporal Heterogeneity of High-Grade Gliomas: From Disease Biology to Therapeutic Implications. Front Oncol 2021; 11:703764. [PMID: 34422657 PMCID: PMC8377724 DOI: 10.3389/fonc.2021.703764] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastomas (GBM) are the most common and aggressive tumors of the central nervous system. Rapid tumor growth and diffuse infiltration into healthy brain tissue, along with high intratumoral heterogeneity, challenge therapeutic efficacy and prognosis. A better understanding of spatiotemporal tumor heterogeneity at the histological, cellular, molecular, and dynamic levels would accelerate the development of novel treatments for this devastating brain cancer. Histologically, GBM is characterized by nuclear atypia, cellular pleomorphism, necrosis, microvascular proliferation, and pseudopalisades. At the cellular level, the glioma microenvironment comprises a heterogeneous landscape of cell populations, including tumor cells, non-transformed/reactive glial and neural cells, immune cells, mesenchymal cells, and stem cells, which support tumor growth and invasion through complex network crosstalk. Genomic and transcriptomic analyses of gliomas have revealed significant inter and intratumoral heterogeneity and insights into their molecular pathogenesis. Moreover, recent evidence suggests that diverse dynamics of collective motion patterns exist in glioma tumors, which correlate with histological features. We hypothesize that glioma heterogeneity is not stochastic, but rather arises from organized and dynamic attributes, which favor glioma malignancy and influences treatment regimens. This review highlights the importance of an integrative approach of glioma histopathological features, single-cell and spatially resolved transcriptomic and cellular dynamics to understand tumor heterogeneity and maximize therapeutic effects.
Collapse
Affiliation(s)
- Andrea Comba
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Todd Hollon
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Wajd N Al-Holou
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yoshie Umemura
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J Nunez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Sebastien Motsch
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, United States
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States.,Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
9
|
Tanshinone IIA sensitizes TRAIL-induced apoptosis in glioblastoma through inducing the expression of death receptors (and suppressing STAT3 activation). Brain Res 2021; 1766:147515. [PMID: 33984327 DOI: 10.1016/j.brainres.2021.147515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/12/2021] [Accepted: 05/06/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE This work was designed to explore whether the combination of Tanshinone IIA (T-IIA) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has a direct anti-cancer effect in glioblastoma (GBM) and the possible mechanisms. METHODS GBM cells (U-87 and U-251 MG) were treated with T-IIA or/and TRAIL, or the expression of death receptors (DRs), DR4 and DR5, was suppressed in GBM cells. The activity of GBM cells was determined by MTT, and the apoptosis was assessed by Hoechst33342 staining and flow cytometry. The expression levels of cleaved caspase-3/8/9, phosphorylated (p)-STAT3 as well as DR4 and DR5 in GBM cells were assessed by Western blotting. A nude mouse xenograft model was constructed to evaluate the effects of T-IIA and TRAIL cotreatment on tumor growth and apoptosis in vivo. RESULTS After T-IIA treatment, GBM cells resumed the sensitivity to TRAIL-induced apoptosis dependent on inhibition of p-STAT3 and activation of DR4, DR5 and caspases. DR4 or/and DR5 knockdown significantly abated the co-effect of T-IIA and TRAIL on GBM cell apoptosis and proliferation. Furthermore, T-IIA and TRAIL cotreatment markedly inhibited the growth of transplanted tumor and activated U87 cell apoptosis in nude mice. CONCLUSION T-IIA increases TRAIL-induced apoptosis by downregulating STAT3 and upregulating DR4 and DR5, indicating T-IIA therapy as a novel treatment strategy for TRAIL-resistant GBM.
Collapse
|
10
|
Sharma A, Bajaj P, Bhandari A, Kaur G. From ayurvedic folk medicine to preclinical neurotherapeutic role of a miraculous herb, Tinospora cordifolia. Neurochem Int 2020; 141:104891. [PMID: 33137454 DOI: 10.1016/j.neuint.2020.104891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 10/23/2020] [Accepted: 10/23/2020] [Indexed: 01/23/2023]
Abstract
In Ayurveda, the age-old Indian traditional system of medicine, health is considered to be achieved as equilibrium of physical and mental wellbeing and brain related ailments are recognized as 'Vatavyadi'. Rasayana herbs were mainly used for pharmacological treatment of neurological diseases and Tinospora cordifolia is one of the popular Rasayana herbs of Ayurveda. The traditional claims of therapeutic activity of this herb for treatment of fever, diabetes, anxiety, immunodeficiency, memory deficit and psychological problems have been explored by different research groups using reverse pharmacology and advance technological approaches. The aim of current review is to compile and discuss the neurotherapeutic potential of T. cordifolia in the light of various preclinical and clinical studies from literature. This review summarizes the information about different extracts of this herb and decoctions used for various neuro-related problems such as neurodegenerative diseases, neuroinflammation, sleep disorders, neural cancers, memory and cognition deficits and psychological problems besides other potential activities. The review also provides the knowledge of underlying therapeutic mechanism of T. cordifolia and its active phytoconstituents.
Collapse
Affiliation(s)
- Anuradha Sharma
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India; Current Address: Mood Disorders and Suicide Research Lab, Department of Psychiatry, University of Illinois at Chicago, Illinois, 60612, USA
| | - Payal Bajaj
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India
| | - Anmol Bhandari
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India
| | - Gurcharan Kaur
- Medical Biotechnology Laboratory, Department of Biotechnology, Guru Nanak Dev University, Amritsar, 143005, India.
| |
Collapse
|
11
|
Birch JL, Coull BJ, Spender LC, Watt C, Willison A, Syed N, Chalmers AJ, Hossain-Ibrahim MK, Inman GJ. Multifaceted transforming growth factor-beta (TGFβ) signalling in glioblastoma. Cell Signal 2020; 72:109638. [PMID: 32320860 DOI: 10.1016/j.cellsig.2020.109638] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) is an aggressive and devastating primary brain cancer which responds very poorly to treatment. The average survival time of patients is only 14-15 months from diagnosis so there is a clear and unmet need for the development of novel targeted therapies to improve patient outcomes. The multifunctional cytokine TGFβ plays fundamental roles in development, adult tissue homeostasis, tissue wound repair and immune responses. Dysfunction of TGFβ signalling has been implicated in both the development and progression of many tumour types including GBM, thereby potentially providing an actionable target for its treatment. This review will examine TGFβ signalling mechanisms and their role in the development and progression of GBM. The targeting of TGFβ signalling using a variety of approaches including the TGFβ binding protein Decorin will be highlighted as attractive therapeutic strategies.
Collapse
Affiliation(s)
| | - Barry J Coull
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Lindsay C Spender
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Courtney Watt
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Alice Willison
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Nelofer Syed
- The John Fulcher Molecular Neuro-Oncology Laboratory, Division of Brain Sciences, Imperial College London, London, UK
| | | | - M Kismet Hossain-Ibrahim
- Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK; Department of Neurosurgery, Ninewells Hospital and Medical School, NHS Tayside, Dundee, UK
| | - Gareth J Inman
- CRUK Beatson Institute, Glasgow, UK; Division of Cellular and Molecular Medicine, School of Medicine, University of Dundee, Dundee, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
12
|
Mathen P, Rowe L, Mackey M, Smart D, Tofilon P, Camphausen K. Radiosensitizers in the temozolomide era for newly diagnosed glioblastoma. Neurooncol Pract 2019; 7:268-276. [PMID: 32537176 DOI: 10.1093/nop/npz057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a challenging diagnosis with almost universally poor prognosis. Though the survival advantage of postoperative radiation (RT) is well established, around 90% of patients will fail in the RT field. The high likelihood of local failure suggests the efficacy of RT needs to be improved to improve clinical outcomes. Radiosensitizers are an established method of enhancing RT cell killing through the addition of a pharmaceutical agent. Though the majority of trials using radiosensitizers have historically been unsuccessful, there continues to be interest with a variety of approaches having been employed. Epidermal growth factor receptor inhibitors, histone deacetylase inhibitors, antiangiogenic agents, and a number of other molecularly targeted agents have all been investigated as potential methods of radiosensitization in the temozolomide era. Outcomes have varied both in terms of toxicity and survival, but some agents such as valproic acid and bortezomib have demonstrated promising results. However, reporting of results in phase 2 trials in newly diagnosed GBM have been inconsistent, with no standard in reporting progression-free survival and toxicity. There is a pressing need for investigation of new agents; however, nearly all phase 3 trials of GBM patients of the past 25 years have demonstrated no improvement in outcomes. One proposed explanation for this is the selection of agents lacking sufficient preclinical data and/or based on poorly designed phase 2 trials. Radiosensitization may represent a viable strategy for improving GBM outcomes in newly diagnosed patients, and further investigation using agents with promising phase 2 data is warranted.
Collapse
Affiliation(s)
- Peter Mathen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Lindsay Rowe
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Megan Mackey
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - DeeDee Smart
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Philip Tofilon
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
13
|
Salaroglio IC, Abate C, Rolando B, Battaglia L, Gazzano E, Colombino E, Costamagna C, Annovazzi L, Mellai M, Berardi F, Capucchio MT, Schiffer D, Riganti C. Validation of Thiosemicarbazone Compounds as P-Glycoprotein Inhibitors in Human Primary Brain-Blood Barrier and Glioblastoma Stem Cells. Mol Pharm 2019; 16:3361-3373. [PMID: 31265310 DOI: 10.1021/acs.molpharmaceut.9b00018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
P-glycoprotein (Pgp) is highly expressed on blood-brain barrier (BBB) and glioblastoma (GB) cells, particularly on cancer stem cells (SC). Pgp recognizes a broad spectrum of substrates, limiting the therapeutic efficacy of several chemotherapeutic drugs in eradicating GB SC. Finding effective and safe inhibitors of Pgp that improve drug delivery across the BBB and target GB SC is open to investigation. We previously identified a series of thiosemicarbazone compounds that inhibit Pgp with an EC50 in the nanomolar range, and herein, we investigate the efficacy of three of them in bypassing Pgp-mediated drug efflux in primary human BBB and GB cells. At 10 nM, the compounds were not cytotoxic for the brain microvascular endothelial hCMEC/D3 cell line, but they markedly enhanced the permeability of the Pgp-substrate doxorubicin through the BBB. Thiosemicarbazone derivatives increased doxorubicin uptake in GB, with greater effects in the Pgp-rich SC clones than in the differentiated clones derived from the same tumor. All compounds increased intratumor doxorubicin accumulation and consequent toxicity in GB growing under competent BBB, producing significant killing of GB SC. The compounds crossed the BBB monolayer. The most stable derivative, 10a, had a half-life in serum of 4.2 h. The coadministration of doxorubicin plus 10a significantly reduced the growth of orthotopic GB-SC xenografts, without eliciting toxic side effects. Our work suggests that the thiosemicarbazone compounds are able to transform doxorubicin, a prototype BBB-impermeable drug, into a BBB-permeable drug. Bypassing Pgp-mediated drug efflux in both BBB and GB SC, thiosemicarbazones might increase the success of chemotherapy in targeting GB SC, which represent the most aggressive and difficult components to eradicate.
Collapse
Affiliation(s)
- Iris Chiara Salaroglio
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco , Università di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco , Università di Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco , Università di Torino , via Pietro Giuria 9 , 10125 Torino , Italy
| | - Elena Gazzano
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Elena Colombino
- Dipartimento di Scienze Veterinarie , Università di Torino , Largo Braccini 2 , 10095 Grugliasco , Italy
| | - Costanzo Costamagna
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Laura Annovazzi
- Centro Ricerche , Fondazione Policlinico di Monza , via Pietro Micca 29 , 13100 Vercelli , Italy
| | - Marta Mellai
- Dipartimento di Scienze della Salute , Università del Piemonte Orientale , corso Mazzini 18 , 28100 Novara , Italy
| | - Francesco Berardi
- Dipartimento di Farmacia-Scienze del Farmaco , Università di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy
| | - Maria Teresa Capucchio
- Dipartimento di Scienze Veterinarie , Università di Torino , Largo Braccini 2 , 10095 Grugliasco , Italy
| | - Davide Schiffer
- Dipartimento di Neuroscienze , Università di Torino , via Cherasco 15 , 10126 Torino , Italy
| | - Chiara Riganti
- Dipartimento di Oncologia , Università di Torino , via Santena 5/bis , 10126 Torino , Italy
| |
Collapse
|
14
|
Serrano-Saenz S, Palacios C, Delgado-Bellido D, López-Jiménez L, Garcia-Diaz A, Soto-Serrano Y, Casal JI, Bartolomé RA, Fernández-Luna JL, López-Rivas A, Oliver FJ. PIM kinases mediate resistance of glioblastoma cells to TRAIL by a p62/SQSTM1-dependent mechanism. Cell Death Dis 2019; 10:51. [PMID: 30718520 PMCID: PMC6362213 DOI: 10.1038/s41419-018-1293-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/07/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor and is associated with poor prognosis. GBM cells are frequently resistant to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and finding new combinatorial therapies to sensitize glioma cells to TRAIL remains an important challenge. PIM kinases are serine/threonine kinases that promote cell survival and proliferation and are highly expressed in different tumors. In this work, we studied the role of PIM kinases as regulators of TRAIL sensitivity in GBM cells. Remarkably, PIM inhibition or knockdown facilitated activation by TRAIL of a TRAIL-R2/DR5-mediated and mitochondria-operated apoptotic pathway in TRAIL-resistant GBM cells. The sensitizing effect of PIM knockdown on TRAIL-induced apoptosis was mediated by enhanced caspase-8 recruitment to and activation at the death-inducing signaling complex (DISC). Interestingly, TRAIL-induced internalization of TRAIL-R2/DR5 was significantly reduced in PIM knockdown cells. Phospho-proteome profiling revealed a decreased phosphorylation of p62/SQSTM1 after PIM knockdown. Our results also showed an interaction between p62/SQSTM1 and the DISC that was reverted after PIM knockdown. In line with this, p62/SQSTM1 ablation increased TRAIL-R2/DR5 levels and facilitated TRAIL-induced caspase-8 activation, revealing an inhibitory role of p62/SQSTM1 in TRAIL-mediated apoptosis in GBM. Conversely, upregulation of TRAIL-R2/DR5 upon PIM inhibition and apoptosis induced by the combination of PIM inhibitor and TRAIL were abrogated by a constitutively phosphorylated p62/SQSTM1S332E mutant. Globally, our data represent the first evidence that PIM kinases regulate TRAIL-induced apoptosis in GBM and identify a specific role of p62/SQSTM1Ser332 phosphorylation in the regulation of the extrinsic apoptosis pathway activated by TRAIL.
Collapse
Affiliation(s)
- Santiago Serrano-Saenz
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain.,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain
| | - Carmen Palacios
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain.,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, CIBERONC, Avda Américo Vespucio 24, 41092, Sevilla, Spain
| | - Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Laura López-Jiménez
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - Yolanda Soto-Serrano
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain
| | - J Ignacio Casal
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039, Madrid, Spain
| | - Rubén A Bartolomé
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28039, Madrid, Spain
| | - José Luis Fernández-Luna
- HUMV-Hospital Universitario Marqués de Valdecilla Avenida Valdecilla, 25, 39008, Santander, Cantabria, Spain
| | - Abelardo López-Rivas
- Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain. .,Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, CSIC-Universidad de Sevilla-Universidad Pablo de Olavide, CIBERONC, Avda Américo Vespucio 24, 41092, Sevilla, Spain.
| | - F Javier Oliver
- Instituto de Parasitología y Biomedicina López-Neyra, CSIC, CIBERONC, Parque Tecnológico Ciencias de la Salud, Avenida del Conocimiento, s/n, 18100, Armilla, Granada, Spain. .,Centro de Investigación Biomédica en Red-Oncología (CIBERONC), Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
15
|
Jantas D, Grygier B, Gołda S, Chwastek J, Zatorska J, Tertil M. An endogenous and ectopic expression of metabotropic glutamate receptor 8 (mGluR8) inhibits proliferation and increases chemosensitivity of human neuroblastoma and glioma cells. Cancer Lett 2018; 432:1-16. [DOI: 10.1016/j.canlet.2018.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/06/2018] [Accepted: 06/03/2018] [Indexed: 12/11/2022]
|
16
|
Salaroglio IC, Gazzano E, Kopecka J, Chegaev K, Costamagna C, Fruttero R, Guglielmo S, Riganti C. New Tetrahydroisoquinoline Derivatives Overcome Pgp Activity in Brain-Blood Barrier and Glioblastoma Multiforme in Vitro. Molecules 2018; 23:molecules23061401. [PMID: 29890725 PMCID: PMC6099747 DOI: 10.3390/molecules23061401] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 11/16/2022] Open
Abstract
P-glycoprotein (Pgp) determines resistance to a broad spectrum of drugs used against glioblastoma multiforme (GB). Indeed, Pgp is highly expressed in GB stem cells and in the brain-blood barrier (BBB), the peculiar endothelium surrounding the brain. Inhibiting Pgp activity in the BBB and GB is still an open challenge. Here, we tested the efficacy of a small library of tetrahydroisoquinoline derivatives with an EC50 for Pgp ≤ 50 nM, in primary human BBB cells and in patient-derived GB samples, from which we isolated differentiated/adherent cells (AC, i.e., Pgp-negative/doxorubicin-sensitive cells) and stem cells (neurospheres, NS, i.e., Pgp-positive/doxorubicin-resistant cells). Three compounds used at 1 nM increased the delivery of doxorubicin, a typical substrate of Pgp, across BBB monolayer, without altering the expression and activity of other transporters. The compounds increased the drug accumulation within NS, restoring doxorubicin-induced necrosis and apoptosis, and reducing cell viability. In co-culture systems, the compounds added to the luminal face of BBB increased the delivery of doxorubicin to NS growing under BBB and rescued the drug’s cytotoxicity. Our work identified new ligands of Pgp active at low nanomolar concentrations. These compounds reduce Pgp activity in BBB and GB and improve in vitro chemotherapy efficacy in this tumor.
Collapse
Affiliation(s)
| | - Elena Gazzano
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Joanna Kopecka
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Costanzo Costamagna
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| | - Roberta Fruttero
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Stefano Guglielmo
- Department of Drug Science and Technology, University of Torino, via Pietro Giuria 9, 10125, Torino, Italy.
| | - Chiara Riganti
- Department of Oncology, University of Torino, via Santena 5/bis, 10126, Torino Italy.
| |
Collapse
|
17
|
Nordling-David MM, Yaffe R, Guez D, Meirow H, Last D, Grad E, Salomon S, Sharabi S, Levi-Kalisman Y, Golomb G, Mardor Y. Liposomal temozolomide drug delivery using convection enhanced delivery. J Control Release 2017; 261:138-146. [DOI: 10.1016/j.jconrel.2017.06.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/11/2022]
|
18
|
Bianco J, Bastiancich C, Jankovski A, des Rieux A, Préat V, Danhier F. On glioblastoma and the search for a cure: where do we stand? Cell Mol Life Sci 2017; 74:2451-2466. [PMID: 28210785 PMCID: PMC11107640 DOI: 10.1007/s00018-017-2483-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 01/30/2017] [Indexed: 01/25/2023]
Abstract
Although brain tumours have been documented and recorded since the nineteenth century, 2016 marked 90 years since Percival Bailey and Harvey Cushing coined the term "glioblastoma multiforme". Since that time, although extensive developments in diagnosis and treatment have been made, relatively little improvement on prognosis has been achieved. The resilience of GBM thus makes treating this tumour one of the biggest challenges currently faced by neuro-oncology. Aggressive and robust development, coupled with difficulties of complete resection, drug delivery and therapeutic resistance to treatment are some of the main issues that this nemesis presents today. Current treatments are far from satisfactory with poor prognosis, and focus on palliative management rather than curative intervention. However, therapeutic research leading to developments in novel treatment stratagems show promise in combating this disease. Here we present a review on GBM, looking at the history and advances which have shaped neurosurgery over the last century that cumulate to the present day management of GBM, while also exploring future perspectives in treatment options that could lead to new treatments on the road to a cure.
Collapse
Affiliation(s)
- John Bianco
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier 73, bte B1 73.12, 1200, Brussels, Belgium.
| | - Chiara Bastiancich
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier 73, bte B1 73.12, 1200, Brussels, Belgium
| | - Aleksander Jankovski
- Institute of Neuroscience, Université catholique de Louvain, Avenue Hippocrate B1.54.10, 1200, Brussels, Belgium
- Department of Neurosurgery, CHU UCL Namur, Avenue G. Thérasse 1, 5530, Yvoir, Belgium
| | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier 73, bte B1 73.12, 1200, Brussels, Belgium
- Institute of Condensed Matter and Nanosciences, Université catholique de Louvain, 1348, Louvain-la-Neuve, Belgium
| | - Véronique Préat
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier 73, bte B1 73.12, 1200, Brussels, Belgium.
| | - Fabienne Danhier
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, Avenue Mounier 73, bte B1 73.12, 1200, Brussels, Belgium
| |
Collapse
|
19
|
King AR, Corso CD, Chen EM, Song E, Bongiorni P, Chen Z, Sundaram RK, Bindra RS, Saltzman WM. Local DNA Repair Inhibition for Sustained Radiosensitization of High-Grade Gliomas. Mol Cancer Ther 2017; 16:1456-1469. [PMID: 28566437 DOI: 10.1158/1535-7163.mct-16-0788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/14/2017] [Accepted: 05/16/2017] [Indexed: 11/16/2022]
Abstract
High-grade gliomas, such as glioblastoma (GBM) and diffuse intrinsic pontine glioma (DIPG), are characterized by an aggressive phenotype with nearly universal local disease progression despite multimodal treatment, which typically includes chemotherapy, radiotherapy, and possibly surgery. Radiosensitizers that have improved the effects of radiotherapy for extracranial tumors have been ineffective for the treatment of GBM and DIPG, in part due to poor blood-brain barrier penetration and rapid intracranial clearance of small molecules. Here, we demonstrate that nanoparticles can provide sustained drug release and minimal toxicity. When administered locally, these nanoparticles conferred radiosensitization in vitro and improved survival in rats with intracranial gliomas when delivered concurrently with a 5-day course of fractionated radiotherapy. Compared with previous work using locally delivered radiosensitizers and cranial radiation, our approach, based on the rational selection of agents and a clinically relevant radiation dosing schedule, produces the strongest synergistic effects between chemo- and radiotherapy approaches to the treatment of high-grade gliomas. Mol Cancer Ther; 16(8); 1456-69. ©2017 AACR.
Collapse
Affiliation(s)
- Amanda R King
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Christopher D Corso
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Evan M Chen
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Eric Song
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut
| | - Paul Bongiorni
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Zhe Chen
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjini K Sundaram
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ranjit S Bindra
- Department of Therapeutic Radiology, Yale University School of Medicine, New Haven, Connecticut. .,Department of Experimental Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut.
| |
Collapse
|
20
|
Kim M, Kotas J, Rockhill J, Phillips M. A Feasibility Study of Personalized Prescription Schemes for Glioblastoma Patients Using a Proliferation and Invasion Glioma Model. Cancers (Basel) 2017; 9:cancers9050051. [PMID: 28505072 PMCID: PMC5447961 DOI: 10.3390/cancers9050051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 05/09/2017] [Accepted: 05/11/2017] [Indexed: 12/25/2022] Open
Abstract
Purpose: This study investigates the feasibility of personalizing radiotherapy prescription schemes (treatment margins and fractional doses) for glioblastoma (GBM) patients and their potential benefits using a proliferation and invasion (PI) glioma model on phantoms. Methods and Materials: We propose a strategy to personalize radiotherapy prescription schemes by simulating the proliferation and invasion of the tumor in 2D according to the PI glioma model. We demonstrate the strategy and its potential benefits by presenting virtual cases, where the standard and personalized prescriptions were applied to the tumor. Standard prescription was assumed to deliver 46 Gy in 23 fractions to the initial, gross tumor volume (GTV1) plus a 2 cm margin and an additional 14 Gy in 7 fractions to the boost GTV2 plus a 2 cm margin. The virtual cases include the tumors with a moving velocity of 0.029 (slow-move), 0.079 (average-move), and 0.13 (fast-move) mm/day for the gross tumor volume (GTV) with a radius of 1 (small) and 2 (large) cm. For each tumor size and velocity, the margin around GTV1 and GTV2 was varied between 0–6 cm and 1–3 cm, respectively. Equivalent uniform dose (EUD) to normal brain was constrained to the EUD value obtained by using the standard prescription. Various linear dose policies, where the fractional dose is linearly decreasing, constant, or increasing, were investigated to estimate the temporal effect of the radiation dose on tumor cell-kills. The goal was to find the combination of margins for GTV1 and GTV2 and a linear dose policy, which minimize the tumor cell-surviving fraction (SF) under a normal tissue constraint. The efficacy of a personalized prescription was evaluated by tumor EUD and the estimated survival time. Results: The personalized prescription for the slow-move tumors was to use 3.0–3.5 cm margins for GTV1, and a 1.5 cm margin for GTV2. For the average- and fast-move tumors, it was optimal to use a 6.0 cm margin for GTV1 and then 1.5–3.0 cm margins for GTV2, suggesting a course of whole brain therapy followed by a boost to a smaller volume. It was more effective to deliver the boost sequentially using a linearly decreasing fractional dose for all tumors. Personalized prescriptions led to surviving fractions of 0.001–0.465% compared to the standard prescription, and increased the tumor EUDs by 25.3–49.3% and estimated survival times by 7.6–22.2 months. Conclusions: Personalizing treatment margins based on the measured proliferative capacity of GBM tumor cells can potentially lead to significant improvements in tumor cell kill and related clinical outcomes.
Collapse
Affiliation(s)
- Minsun Kim
- Department of Radiation Oncology, University of Washington, Seattle, WA 98195, USA.
| | - Jakob Kotas
- Department of Mathematics, University of Portland, Portland, OR 97203, USA.
| | - Jason Rockhill
- Department of Radiation Oncology, University of Washington, Seattle, WA 98195, USA.
| | - Mark Phillips
- Department of Radiation Oncology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
21
|
Management of GBM: a problem of local recurrence. J Neurooncol 2017; 134:487-493. [PMID: 28378194 DOI: 10.1007/s11060-016-2347-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 12/23/2016] [Indexed: 01/22/2023]
Abstract
Forty years ago, adjuvant treatment of patients with GBM using fractionated radiotherapy following surgery was shown to substantially improve survival compared to surgery alone. However, even with the addition of temozolomide to radiotherapy, overall survival is quite limited and local failure remains a fundamental problem, despite multiple attempts to increase dose to the tumor target. This review presents the historical background and clinical rationale leading to the current standard of care consisting of 60 Gy total dose in 2 Gy fractions to the MRI-defined targets in younger, high performance status patients and more hypofractionated regimens in elderly and/or debilitated patients. Particle therapies offer the potential to increase local control while reducing dose and, potentially, long-term neurocognitive toxicity. However, improvements in systemic therapies for GBM will need to be implemented before the full benefits of improved local control can be realized.
Collapse
|
22
|
Timbie KF, Afzal U, Date A, Zhang C, Song J, Wilson Miller G, Suk JS, Hanes J, Price RJ. MR image-guided delivery of cisplatin-loaded brain-penetrating nanoparticles to invasive glioma with focused ultrasound. J Control Release 2017; 263:120-131. [PMID: 28288892 DOI: 10.1016/j.jconrel.2017.03.017] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 02/21/2017] [Accepted: 03/08/2017] [Indexed: 12/24/2022]
Abstract
Systemically administered chemotherapeutic drugs are often ineffective in the treatment of invasive brain tumors due to poor therapeutic index. Within gliomas, despite the presence of heterogeneously leaky microvessels, dense extracellular matrix and high interstitial pressure generate a "blood-tumor barrier" (BTB), which inhibits drug delivery and distribution. Meanwhile, beyond the contrast MRI-enhancing edge of the tumor, invasive cancer cells are protected by the intact blood-brain barrier (BBB). Here, we tested whether brain-penetrating nanoparticles (BPN) that possess dense surface coatings of polyethylene glycol (PEG) and are loaded with cisplatin (CDDP) could be delivered across both the blood-tumor and blood-brain barriers with MR image-guided focused ultrasound (MRgFUS), and whether this treatment could control glioma growth and invasiveness. To this end, we first established that MRgFUS is capable of significantly enhancing the delivery of ~60nm fluorescent tracer BPN across the blood-tumor barrier in both the 9L (6-fold improvement) gliosarcoma and invasive F98 (28-fold improvement) glioma models. Importantly, BPN delivery across the intact BBB, just beyond the tumor edge, was also markedly increased in both tumor models. We then showed that a CDDP loaded BPN formulation (CDDP-BPN), composed of a blend of polyaspartic acid (PAA) and heavily PEGylated polyaspartic acid (PAA-PEG), was highly stable, provided extended drug release, and was effective against F98 cells in vitro. These CDDP-BPN were delivered from the systemic circulation into orthotopic F98 gliomas using MRgFUS, where they elicited a significant reduction in tumor invasiveness and growth, as well as improved animal survival. We conclude that this therapy may offer a powerful new approach for the treatment invasive gliomas, particularly for preventing and controlling recurrence.
Collapse
Affiliation(s)
- Kelsie F Timbie
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road Building MR5, Charlottesville, VA 22908, United States
| | - Umara Afzal
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Department of Biochemistry, PMAS-Arid Agriculture University, Shamsabad, Muree Road, Rawalpindi, Pakistan
| | - Abhijit Date
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States
| | - Clark Zhang
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States
| | - Ji Song
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road Building MR5, Charlottesville, VA 22908, United States
| | - G Wilson Miller
- Department of Radiology and Medical Imaging, University of Virginia, 480 Ray C Hunt Drive, Charlottesville, VA 22908, United States
| | - Jung Soo Suk
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States
| | - Justin Hanes
- Department of Ophthalmology, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States; Center for Nanomedicine, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Baltimore, MD 21287, United States
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, 415 Lane Road Building MR5, Charlottesville, VA 22908, United States.
| |
Collapse
|
23
|
Ahmed MM, Narendra A, Prasanna P, Coleman CN, Krishnan S. Current Insights in Radiation Combination Therapies: Influence of Omics and Novel Targeted Agents in Defining New Concepts in Radiation Biology and Clinical Radiation Oncology. Semin Radiat Oncol 2016; 26:251-3. [DOI: 10.1016/j.semradonc.2016.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|