1
|
Pan J, Eskandar T, Ahmed Z, Agrawal DK. Biophysical and Biological Mechanisms of Tumor Treating Fields in Glioblastoma. JOURNAL OF CANCER SCIENCE AND CLINICAL THERAPEUTICS 2024; 8:265-270. [PMID: 39364266 PMCID: PMC11448370 DOI: 10.26502/jcsct.5079249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
Glioblastoma (GBM) is one of the most aggressive forms of brain cancer that presents with a median survival rate of 14-30 months and along with a discouraging five-year survival rate of 4-5%. Standard treatment of newly diagnosed GBM, also known as the Stupp protocol, includes a maximally safe surgical resection followed by radiation and chemotherapy. Despite these treatment regimens, recurrence is almost inevitable, emphasizing the need for new therapies to combat the aggressive nature of GBMs. Tumor Treating Fields (TTFs) are a relatively new application to the treatment of GBMs, and results have been promising with both progression-free survival and overall survival when TTFs have been used in combination with temozolomide. This article critically reviews the biophysical and biological mechanisms of TTFs, their clinical efficacy, and discusses the results in clinical trials, including EF-11 and EF-14. Both trials have demonstrated that TTFs can enhance progression free survival and overall survival without compromising quality of life or causing severe adverse effects. Despite the high cost associated with TTFs and the need for further analysis to determine the most effective ways to integrate TTFs into GBM treatments, TTFs represent a significant advancement in GBM therapy and offer hope for improved patient prognosis.
Collapse
Affiliation(s)
- Jeremy Pan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Tony Eskandar
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Zubair Ahmed
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona CA 91766, USA
| |
Collapse
|
2
|
YILDIRIM Z, DOĞAN E, GÜLER KARA H, KOSOVA B, BOZOK V. STING activation increases the efficiency of temozolomide in PTEN harbouring glioblastoma cells. Turk J Med Sci 2024; 54:607-614. [PMID: 39049995 PMCID: PMC11265881 DOI: 10.55730/1300-0144.5828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 06/12/2024] [Accepted: 01/21/2024] [Indexed: 07/27/2024] Open
Abstract
Background/aim Glioblastoma is one of the most aggressive tumours, resistant to all applied therapy regiments and prone to relapse. Median survival rates are therefore only expressed as months. STING agonists are immunomodulatory molecules that activate type I interferon expression, making them potentially useful in regulating the tumour microenvironment. Since PTEN serves as a critical phosphatase in activating interferon-regulating transcription factors and is frequently mutated in glioblastoma cells, this study aimed to investigate STING activation in glioblastoma cell lines, examining whether they harbour the PTEN protein or not.°. Materials and methods T98G and U118MG glioblastoma cell lines were treated with the 2'3'-c-di-AM(PS)2(Rp,Rp) STING agonist together with or without the chemotherapeutic agent temozolomide. cGAS/STING pathway components were subsequently analysed using qRT-PCR, western blot, and ELISA methods. Results Our results showed that PTEN-harbouring T98G cells responded well to STING activation, leading to increased temozolomide efficacy. In contrast, STING activation in U118MG cells did not affect the response to temozolomide. mRNA expression levels of STING, IRF3, NF-KB, and RELA genes were significantly increased at the combined treatment groups in T98G cell line. Conversely, combined treatment with STING agonist and temozolomide did not affect mRNA expression levels of cGAS/STING pathway genes in U118MG cells. Conclusion Our data offers new evidence suggesting that STING agonists can effectively be used to increase temozolomide response in the presence of PTEN protein. Therefore, increased GBM therapy success rates can be achieved by employing the PTEN expression status as a predictive biomarker before treating patients with a chemotherapeutic agent in combination with STING agonist.
Collapse
Affiliation(s)
| | - Eda DOĞAN
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkiye
| | - Hale GÜLER KARA
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkiye
- Department of Medical Biology, Faculty of Medicine, Harran University, Şanlıurfa,
Turkiye
| | - Buket KOSOVA
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkiye
| | - Vildan BOZOK
- Department of Medical Biology, Faculty of Medicine, Ege University, İzmir,
Turkiye
| |
Collapse
|
3
|
Hekimoglu M, Basak AT, Akgun MY, Ozer H, Ozgen U, Maleki R, Saban D, Oktenoğlu T, Ozer AF, Sasani M. The Impact of Extensive Surgical Resection of Butterfly Glioblastomas on Outcomes in the Presence of TERT Mutation and EGFR Amplification: A Retrospective Cohort Study. Cancer Control 2024; 31:10732748241288121. [PMID: 39327682 PMCID: PMC11437567 DOI: 10.1177/10732748241288121] [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: 06/24/2024] [Revised: 09/01/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND AND OBJECTIVES This study aimed to assess if extensive surgical resection enhances outcomes in wild-type Isocitrate Dehydrogenase (IDH) butterfly glioblastoma (B-GBM) patients, despite the presence of Telomerase Reverse Transcriptase (TERT) mutation and Epidermal Growth Factor Receptor (EGFR) amplification. METHODS The study, retrospectively conducted from 2014 to 2022, involved 723 GBM patients, 41 of whom met the criteria for IDH wild-type B-GBM. Exclusion criteria comprised prior diagnoses or treatments for low-grade glial tumors. Surgeons, employing two approaches-partial and extensive surgery-categorized patients based on age, sex, tumor location, corpus callosum involvement, and genetic characteristics. The interval between initial surgery and tumor recurrence/tumor-free period (TR/TFP) and overall survival (OS) were recorded and compared between the partial and extensive resection groups, analyzing the impact of resection width on TR/TFP and OS. Preoperative assessments utilized thin-section cranial computed tomography (CT) and contrast-enhanced magnetic resonance imaging (MRI). Intraoperatively, tumor excision was guided by sodium fluorescein, and margins were delineated via neuronavigation. Genetic alterations (TERT mutations and EGFR amplifications) were correlated with surgical type, TR/TFP, and OS. Karnofsky Performance Scale (KPS) evaluations were performed pre- and post-operatively and at key intervals, comparing outcomes between surgical groups. Standard radiotherapy and chemotherapy regimens were administered to all patients. RESULTS Extensive resection yielded significantly longer TR/TFP compared to partial resection, despite TERT gene mutation and EGFR amplification being linked to shorter TR/TFP and OS. Its impact on OS, however, was not significant. KPS scores indicated a superior quality of life after extensive resection, with sustained improvement upon recurrence. CONCLUSIONS Extensive resection of B-GBM, even in the presence of adverse genetic alterations, may prolong TR/TFP, offering patients a period of improved comfort with minimal distress.
Collapse
Affiliation(s)
- Mehdi Hekimoglu
- Department of Neurosurgery,American Hospital, Istanbul, Turkey
| | | | | | - Hıdır Ozer
- Department of Neurosurgery, Ordu University School of Medicine, Ordu, Turkey
| | - Utku Ozgen
- Department of Neurosurgery,American Hospital, Istanbul, Turkey
| | - Raha Maleki
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, İran
| | - Dina Saban
- Medical Student, Wayne State University School of Medicine, Detroit, MI, USA
| | - Tunc Oktenoğlu
- Department of Neurosurgery,American Hospital, Istanbul, Turkey
- Department of Neurosurgery, Koc University Hospital, Istanbul, Turkey
| | - Ali Fahir Ozer
- Department of Neurosurgery,American Hospital, Istanbul, Turkey
- Department of Neurosurgery, Koc University Hospital, Istanbul, Turkey
| | - Mehdi Sasani
- Department of Neurosurgery,American Hospital, Istanbul, Turkey
- Department of Neurosurgery, Koc University Hospital, Istanbul, Turkey
| |
Collapse
|
4
|
Singh S, Joshi V, Upadhyay A. Amyloids and brain cancer: molecular linkages and crossovers. Biosci Rep 2023; 43:BSR20230489. [PMID: 37335084 PMCID: PMC10548166 DOI: 10.1042/bsr20230489] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
Amyloids are high-order proteinaceous formations deposited in both intra- and extracellular spaces. These aggregates have tendencies to deregulate cellular physiology in multiple ways; for example, altered metabolism, mitochondrial dysfunctions, immune modulation, etc. When amyloids are formed in brain tissues, the endpoint often is death of neurons. However, interesting but least understood is a close connection of amyloids with another set of conditions in which brain cells proliferate at an extraordinary rate and form tumor inside brain. Glioblastoma is one such condition. Increasing number of evidence indicate a possible link between amyloid formation and depositions in brain tumors. Several proteins associated with cell cycle regulation and apoptotic pathways themselves have shown to possess high tendencies to form amyloids. Tumor suppressor protein p53 is one prominent example that mutate, oligomerize and form amyloids leading to loss- or gain-of-functions and cause increased cell proliferation and malignancies. In this review article, we present available examples, genetic links and common pathways that indicate that possibly the two distantly placed pathways: amyloid formation and developing cancers in the brain have similarities and are mechanistically intertwined together.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| | - Vibhuti Joshi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh 201310, India
| | - Arun Upadhyay
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| |
Collapse
|
5
|
Eltoukhy M, Kandula V, Joseph S, Albanese E, Giridharan S. Should Redo Surgery be Offered to Patients with Relapsed Glioblastoma? - Outcome Analyses of a Single Institution Comparative Cohort Study. World Neurosurg 2023; 176:e543-e547. [PMID: 37268188 DOI: 10.1016/j.wneu.2023.05.097] [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: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/04/2023]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the predominant malignant brain tumor originating intracranially. The established first-line treatment postsurgery is concurrent chemoradiation as a definitive measure. However, recurrent GBM's pose a challenge for clinicians who rely on institutional experience to determine the most suitable course of action. Second-line chemotherapy may be administered with or without surgery depending on the institution's practice. This study aims to present our tertiary center institution's experience with recurrent GBM patients who underwent redo surgery. METHODS In this retrospective study we analyzed the surgical and oncological data of patients with recurrent GBM who underwent redo surgery at the Royal Stoke University Hospitals between 2006 and 2015. The group 1 (G1) comprised the reviewed patients, while a control group (G2) was randomly selected, matching the reviewed group by age, primary treatment, and progression-free survival (PFS). The study collected data on various parameters, including overall survival, PFS, extent of surgical resection, and postoperative complications. RESULTS This retrospective study included 30 patients in G1 and 32 patients in G2, matched based on age, primary treatment, and PFS. The study found that the overall survival for the G1 group from the time of first diagnosis was 109 weeks (45-180) compared to 57 weeks (28-127) in the G2 group. The incidence of postoperative complications after the second surgery was 57%, which included hemorrhage, infarction, worsening neurology due to edema, cerebrospinal fluid leak, and wound infection. Furthermore, 50% of the patients in the G1 group who underwent redo surgery received second-line chemotherapy. CONCLUSIONS Our study found that redo surgery for recurrent GBM is a viable treatment option for a select group of patients with good performance status, longer PFS from primary treatment, and compressive symptoms. However, the use of redo surgery varies depending on the institution. A well-designed randomized controlled trial in this population would help establish the standard of surgical care.
Collapse
Affiliation(s)
| | - Viswapathi Kandula
- Department of Neurosurgery, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | - Shibu Joseph
- Department of Radiation Oncology, Riverina Cancer Care Centre, New South Wales, Australia
| | - Erminia Albanese
- Department of Neurosurgery, University Hospitals of North Midlands NHS Trust, Stoke-on-Trent, United Kingdom
| | | |
Collapse
|
6
|
Lin FH, Hsu YC, Chang KC, Shyong YJ. Porous hydroxyapatite carrier enables localized and sustained delivery of honokiol for glioma treatment. Eur J Pharm Biopharm 2023:S0939-6411(23)00169-8. [PMID: 37391090 DOI: 10.1016/j.ejpb.2023.06.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/13/2023] [Accepted: 06/27/2023] [Indexed: 07/02/2023]
Abstract
The objective of this study is to develop hydroxyapatite (HAp) particles for targeted delivery of honokiol to tumor sites after glioma surgical management. Honokiol is released from the HAp-honokiol particles inside cancer cells through endocytosis and subsequent acid lysosomal dissolution. HAp is synthesized using a co-precipitation method, and egg white is added to create porous structures. The HAp is then surface-modified with stearic acid to enhance its hydrophobicity and loaded with honokiol to form HAp-honokiol particles. The synthesized particles are of appropriate size and characteristics for cancer cell uptake. Honokiol remains attached on to the HAp particles in neutral environments due to its hydrophobic nature, but undergoes rapid burst release in acidic environments such as lysosomes. The HAp-honokiol treatment shows a delayed effect on cell viability and cytotoxicity, indicating sustained drug release without compromising drug efficacy. Flow cytometry analysis demonstrates the apoptosis pathway induced by HAp-honokiol in ALTS1C1 glioma cells. In an in vivo study using a mouse glioma model, MRI results showed a 40% reduction in tumor size after HAp-honokiol treatment. These findings suggest that HAp-honokiol particles have potential as an effective drug delivery system for the treatment of glioma.
Collapse
Affiliation(s)
- Feng-Huei Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Hsu
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chi Chang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei, Taiwan
| | - Yan-Jye Shyong
- School of Pharmacy and Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan City, Taiwan.
| |
Collapse
|
7
|
Msheik A, Fares Y, Hamoud M, Atat R. Unraveling the Complexity of Multicentric Gliomas: Insights Into Chronicity and Genetic Aberrations. Cureus 2023; 15:e37284. [PMID: 37168179 PMCID: PMC10165940 DOI: 10.7759/cureus.37284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2023] [Indexed: 05/13/2023] Open
Abstract
Gliomas are among the most common primary tumors of the brain. Discrimination among tumors of more than one focus has segregated the latter into two groups: multifocal gliomas and multicentric gliomas (MCGs). In this case series, outcomes among three patients are described and discussed in light of the findings present in the literature. Ideally, it is crucial to consider genetic testing for categorizing each tumor. This can help determine the original genetic mutations of MCGs and allow to establish necessary screening testing for early detection. We present the cases of three patients diagnosed with cranial gliomas. The first case showed two synchronous gliomas at different loci in the right hemisphere. The second patient showed synchronous lesions on cranial magnetic resonance imaging in each hemisphere. The third case was of a patient with metachronous lesions appearing at different times with similar radiological findings at different loci of the same hemisphere. Discrimination among multifocal and multicentric gliomas requires genetic workup because radiological and temporal findings may fail to allow adequate discrimination.
Collapse
Affiliation(s)
- Ali Msheik
- Neurosurgery, Al Zahraa Hospital University Medical Center, Beirut, LBN
| | - Youssef Fares
- Neurosurgery, Al Zahraa Hospital University Medical Center, Beirut, LBN
| | - Maarouf Hamoud
- Neurosurgery, Al Zahraa Hospital University Medical Center, Beirut, LBN
| | - Rami Atat
- Neurology Division, Faculty of Medicine, Lebanese University, Al Zahraa Hospital University Medical Center, Beirut, LBN
| |
Collapse
|
8
|
Cepeda S, Luppino LT, Pérez-Núñez A, Solheim O, García-García S, Velasco-Casares M, Karlberg A, Eikenes L, Sarabia R, Arrese I, Zamora T, Gonzalez P, Jiménez-Roldán L, Kuttner S. Predicting Regions of Local Recurrence in Glioblastomas Using Voxel-Based Radiomic Features of Multiparametric Postoperative MRI. Cancers (Basel) 2023; 15:1894. [PMID: 36980783 PMCID: PMC10047582 DOI: 10.3390/cancers15061894] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The globally accepted surgical strategy in glioblastomas is removing the enhancing tumor. However, the peritumoral region harbors infiltration areas responsible for future tumor recurrence. This study aimed to evaluate a predictive model that identifies areas of future recurrence using a voxel-based radiomics analysis of magnetic resonance imaging (MRI) data. This multi-institutional study included a retrospective analysis of patients diagnosed with glioblastoma who underwent surgery with complete resection of the enhancing tumor. Fifty-five patients met the selection criteria. The study sample was split into training (N = 40) and testing (N = 15) datasets. Follow-up MRI was used for ground truth definition, and postoperative structural multiparametric MRI was used to extract voxel-based radiomic features. Deformable coregistration was used to register the MRI sequences for each patient, followed by segmentation of the peritumoral region in the postoperative scan and the enhancing tumor in the follow-up scan. Peritumoral voxels overlapping with enhancing tumor voxels were labeled as recurrence, while non-overlapping voxels were labeled as nonrecurrence. Voxel-based radiomic features were extracted from the peritumoral region. Four machine learning-based classifiers were trained for recurrence prediction. A region-based evaluation approach was used for model evaluation. The Categorical Boosting (CatBoost) classifier obtained the best performance on the testing dataset with an average area under the curve (AUC) of 0.81 ± 0.09 and an accuracy of 0.84 ± 0.06, using region-based evaluation. There was a clear visual correspondence between predicted and actual recurrence regions. We have developed a method that accurately predicts the region of future tumor recurrence in MRI scans of glioblastoma patients. This could enable the adaptation of surgical and radiotherapy treatment to these areas to potentially prolong the survival of these patients.
Collapse
Affiliation(s)
- Santiago Cepeda
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Luigi Tommaso Luppino
- Department of Physics and Technology, UiT The Arctic University of Norway, 9019 Tromsø, Norway
| | - Angel Pérez-Núñez
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Ole Solheim
- Department of Neurosurgery, St. Olavs University Hospital, 7030 Trondheim, Norway
- Department of Neuromedicine and Movement Science, Norwegian University of Science and Technology, 7034 Trondheim, Norway
| | - Sergio García-García
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | | | - Anna Karlberg
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
- Department of Radiology and Nuclear Medicine, St. Olavs Hospital, Trondheim University Hospital, 7030 Trondheim, Norway
| | - Live Eikenes
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Rosario Sarabia
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Ignacio Arrese
- Department of Neurosurgery, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Tomás Zamora
- Department of Pathology, Río Hortega University Hospital, 47014 Valladolid, Spain
| | - Pedro Gonzalez
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Luis Jiménez-Roldán
- Department of Neurosurgery, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
- Department of Surgery, School of Medicine, Complutense University, 28040 Madrid, Spain
- Instituto de Investigación Sanitaria, 12 de Octubre University Hospital (i+12), 28041 Madrid, Spain
| | - Samuel Kuttner
- Department of Physics and Technology, UiT The Arctic University of Norway, 9019 Tromsø, Norway
- The PET Imaging Center, University Hospital of North Norway, 9019 Tromsø, Norway
| |
Collapse
|
9
|
Stem cell-nanomedicine system as a theranostic bio-gadolinium agent for targeted neutron capture cancer therapy. Nat Commun 2023; 14:285. [PMID: 36650171 PMCID: PMC9845336 DOI: 10.1038/s41467-023-35935-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
The potential clinical application of gadolinium-neutron capture therapy (Gd-NCT) for glioblastoma multiforme (GBM) treatment has been compromised by the fast clearance and nonspecific biodistribution of gadolinium-based agents. We have developed a stem cell-nanoparticle system (SNS) to actively target GBM for advanced Gd-NCT by magnetizing umbilical cord mesenchymal stem cells (UMSCs) using gadodiamide-concealed magnetic nanoparticles (Gd-FPFNP). Nanoformulated gadodiamide shielded by a dense surface composed of fucoidan and polyvinyl alcohol demonstrates enhanced cellular association and biocompatibility in UMSCs. The SNS preserves the ability of UMSCs to actively penetrate the blood brain barrier and home to GBM and, when magnetically navigates by an external magnetic field, an 8-fold increase in tumor-to-blood ratio is achieved compared with clinical data. In an orthotopic GBM-bearing rat model, using a single dose of irradiation and an ultra-low gadolinium dose (200 μg kg-1), SNS significantly attenuates GBM progression without inducing safety issues, prolonging median survival 2.5-fold compared to free gadodiamide. The SNS is a cell-based delivery system that integrates the strengths of cell therapy and nanotechnology, which provides an alternative strategy for the treatment of brain diseases.
Collapse
|
10
|
Acharekar A, Bachal K, Shirke P, Thorat R, Banerjee A, Gardi N, Majumder A, Dutt S. Substrate stiffness regulates the recurrent glioblastoma cell morphology and aggressiveness. Matrix Biol 2023; 115:107-127. [PMID: 36563706 DOI: 10.1016/j.matbio.2022.12.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/25/2022]
Abstract
Recurrent glioblastoma is highly aggressive with currently no specific treatment regime. Therefore, to identify novel therapeutic targets for recurrent GBM, we used a cellular model developed in our lab from commercially available cell line U87MG and patient-derived cultures that allows the comparison between radiation naïve (Parent) and recurrent GBM cells generated after parent cells are exposed to lethal dose of radiation. Total RNA-seq of parent and recurrent population revealed significant upregulation of cell-ECM interactions pathway in the recurrent population. These results led us to hypothesize that the physical microenvironment contributes to the aggressiveness of recurrent GBM. To verify this, we cultured parent and recurrent GBM cells on collagen-coated polyacrylamide gels mimicking the stiffness of normal brain (Young's modulus E = 0.5kPa) or tumorigenic brain (E = 10kPa) and tissue culture plastic dishes (E ∼ 1 GPa). We found that compared to parent cells, recurrent cells showed higher proliferation, invasion, migration, and resistance to EGFR inhibitor. Using orthotopic GBM mouse model and resection model, we demonstrate that recurrent cells cultured on 0.5kPa had higher in vivo tumorigenicity and recurrent disease progression than parent cells, whereas these differences were insignificant when parent and recurrent cells were cultured on plastic substrates. Furthermore, recurrent cells on 0.5kPa showed high expression of ECM proteins like Collagen, MMP2 and MMP9. These proteins were also significantly upregulated in recurrent patient biopsies. Additionally, the brain of mice injected with recurrent cells grown on 0.5kPa showed higher Young's moduli suggesting the ability of these cells to make the surrounding ECM stiffer. Total RNA-seq of parent and recurrent cells grown on plastic and 0.5kpa identified PLEKHA7 significantly upregulated specifically in recurrent cells grown on 0.5 kPa substrate. PLEKHA7 was also found to be high in recurrent GBM patient biopsies. Accordingly, PLEKHA7 knockdown reduced invasion and survival of recurrent GBM cells. Together, these data provide an in vitro model system that captures the observed in vivo and clinical behavior of recurrent GBM by mimicking mechanical microenvironment and identifies PLEKHA7 as a novel potential target for recurrent GBM.
Collapse
Affiliation(s)
- Anagha Acharekar
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Ketaki Bachal
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Pallavi Shirke
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Rahul Thorat
- Laboratory Animal Facility, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre (TMC), Kharghar, Navi Mumbai, India
| | - Archisman Banerjee
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Nilesh Gardi
- Department of Medical Oncology, Tata Memorial Hospital, Tata Memorial Centre, Navi Mumbai, Maharashtra 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India
| | - Abhijit Majumder
- M-Lab, Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India
| | - Shilpee Dutt
- Shilpee Dutt laboratory, Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai, 410210, India.; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400085, India..
| |
Collapse
|
11
|
Lipkova J, Chen RJ, Chen B, Lu MY, Barbieri M, Shao D, Vaidya AJ, Chen C, Zhuang L, Williamson DFK, Shaban M, Chen TY, Mahmood F. Artificial intelligence for multimodal data integration in oncology. Cancer Cell 2022; 40:1095-1110. [PMID: 36220072 PMCID: PMC10655164 DOI: 10.1016/j.ccell.2022.09.012] [Citation(s) in RCA: 201] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/12/2022] [Accepted: 09/15/2022] [Indexed: 02/07/2023]
Abstract
In oncology, the patient state is characterized by a whole spectrum of modalities, ranging from radiology, histology, and genomics to electronic health records. Current artificial intelligence (AI) models operate mainly in the realm of a single modality, neglecting the broader clinical context, which inevitably diminishes their potential. Integration of different data modalities provides opportunities to increase robustness and accuracy of diagnostic and prognostic models, bringing AI closer to clinical practice. AI models are also capable of discovering novel patterns within and across modalities suitable for explaining differences in patient outcomes or treatment resistance. The insights gleaned from such models can guide exploration studies and contribute to the discovery of novel biomarkers and therapeutic targets. To support these advances, here we present a synopsis of AI methods and strategies for multimodal data fusion and association discovery. We outline approaches for AI interpretability and directions for AI-driven exploration through multimodal data interconnections. We examine challenges in clinical adoption and discuss emerging solutions.
Collapse
Affiliation(s)
- Jana Lipkova
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Richard J Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Bowen Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Computer Science, Harvard University, Cambridge, MA, USA
| | - Ming Y Lu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Matteo Barbieri
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel Shao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Health Sciences and Technology (HST), Cambridge, MA, USA
| | - Anurag J Vaidya
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Harvard-MIT Health Sciences and Technology (HST), Cambridge, MA, USA
| | - Chengkuan Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Luoting Zhuang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Drew F K Williamson
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Muhammad Shaban
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tiffany Y Chen
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Faisal Mahmood
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA; Data Science Program, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
12
|
Dajani S, Hill VB, Kalapurakal JA, Horbinski CM, Nesbit EG, Sachdev S, Yalamanchili A, Thomas TO. Imaging of GBM in the Age of Molecular Markers and MRI Guided Adaptive Radiation Therapy. J Clin Med 2022; 11:jcm11195961. [PMID: 36233828 PMCID: PMC9572863 DOI: 10.3390/jcm11195961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 09/26/2022] [Accepted: 09/29/2022] [Indexed: 12/03/2022] Open
Abstract
Glioblastoma (GBM) continues to be one of the most lethal malignancies and is almost always fatal. In this review article, the role of radiation therapy, systemic therapy, as well as the molecular basis of classifying GBM is described. Technological advances in the treatment of GBM are outlined as well as the diagnostic imaging characteristics of this tumor. In addition, factors that affect prognosis such as differentiating progression from treatment effect is discussed. The role of MRI guided radiation therapy and how this technology may provide a mechanism to improve the care of patients with this disease are described.
Collapse
|
13
|
Wang X, Fan L, Wang X, Luo T, Liu L. Cyclophilin A contributes to shikonin-induced glioma cell necroptosis and promotion of chromatinolysis. Sci Rep 2022; 12:14675. [PMID: 36038617 PMCID: PMC9424531 DOI: 10.1038/s41598-022-19066-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
Shikonin induces glioma cell death via necroptosis, a caspase-independent programmed cell death pathway that is chiefly regulated by receptor-interacting serine/threonine protein kinase1 (RIP1) and 3 (RIP3). Chromatinolysis is considered as one of the key events leading to cell death during necroptosis. It is usually accompanied with nuclear translocation of AIF and formation of γ-H2AX. Cyclophilin A (CypA) is reported to participate in the nuclear translocation of AIF during apoptosis. However, it remains unclear whether CypA contributes to necroptosis and regulation of chromatinolysis. In this study, our results revealed for the first time that shikonin promoted time-dependent CypA activation, which contributed to nuclear translocation of AIF and γ-H2AX formation. In vitro studies showed that knockdown of CypA by siRNA or inhibition of CypA by its specific inhibitor, cyclosporine A (CsA), not only significantly mitigated shikonin-induced glioma cell death, but also prevented chromatinolysis. Mechanistically, activated CypA targeted mitochondria and triggered mitochondrial superoxide overproduction, which then promoted AIF translocation from mitochondria into the nucleus by depolarizing the mitochondria and intensified the formation of γ-H2AX by promoting intracellular accumulation of ROS. Additionally, the CypA in the nucleus can form DNA degradation complexes with AIF and γ-H2AX, which also promote the execution of chromatinolysis. Thus, we demonstrate that CypA contributes to shikonin-induced glioma cell necroptosis and promotion of chromatinolysis.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, China
| | - Liwen Fan
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xuanzhong Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Tianfei Luo
- Department of Neurology, First Hospital of Jilin University, Changchun, China.
| | - Linlin Liu
- Department of Radiotherapy, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
14
|
Sareen H, Ma Y, Becker TM, Roberts TL, de Souza P, Powter B. Molecular Biomarkers in Glioblastoma: A Systematic Review and Meta-Analysis. Int J Mol Sci 2022; 23:ijms23168835. [PMID: 36012105 PMCID: PMC9408540 DOI: 10.3390/ijms23168835] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Glioblastoma (GBM) is a highly aggressive cancer with poor prognosis that needs better treatment modalities. Moreover, there is a lack of reliable biomarkers to predict the response and outcome of current or newly designed therapies. While several molecular markers have been proposed as potential biomarkers for GBM, their uptake into clinical settings is slow and impeded by marker heterogeneity. Detailed assessment of prognostic and predictive value for biomarkers in well-defined clinical trial settings, if available, is scattered throughout the literature. Here we conducted a systematic review and meta-analysis to evaluate the prognostic and predictive significance of clinically relevant molecular biomarkers in GBM patients. Material and methods: A comprehensive literature search was conducted to retrieve publications from 3 databases (Pubmed, Cochrane and Embase) from January 2010 to December 2021, using specific terms. The combined hazard ratios (HR) and confidence intervals (95% CI) were used to evaluate the association of biomarkers with overall survival (OS) in GBM patients. Results: Twenty-six out of 1831 screened articles were included in this review. Nineteen articles were included in the meta-analyses, and 7 articles were quantitatively summarised. Fourteen studies with 1231 GBM patients showed a significant association of MGMT methylation with better OS with the pooled HR of 1.66 (95% CI 1.32−2.09, p < 0.0001, random effect). Five studies including 541 GBM patients analysed for the prognostic significance of IDH1 mutation showed significantly better OS in patients with IDH1 mutation with a pooled HR of 2.37 (95% CI 1.81−3.12; p < 0.00001]. Meta-analysis performed on 5 studies including 575 GBM patients presenting with either amplification or high expression of EGFR gene did not reveal any prognostic significance with a pooled HR of 1.31 (95% CI 0.96−1.79; p = 0.08). Conclusions: MGMT promoter methylation and IDH1 mutation are significantly associated with better OS in GBM patients. No significant associations were found between EGFR amplification or overexpression with OS.
Collapse
Affiliation(s)
- Heena Sareen
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- South-Western Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- Correspondence: ; Tel.: +61-0406937108
| | - Yafeng Ma
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- South-Western Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
| | - Therese M. Becker
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- South-Western Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Tara L. Roberts
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
- South-Western Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
| | - Paul de Souza
- South-Western Clinical School, University of New South Wales, Liverpool, NSW 2170, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW 2560, Australia
- Liverpool Hospital, Liverpool, NSW 2170, Australia
| | - Branka Powter
- Centre for Circulating Tumour Cell Diagnostics and Research, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia
| |
Collapse
|
15
|
Schulz JA, Rodgers LT, Kryscio RJ, Hartz AMS, Bauer B. Characterization and comparison of human glioblastoma models. BMC Cancer 2022; 22:844. [PMID: 35922758 PMCID: PMC9347152 DOI: 10.1186/s12885-022-09910-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
Glioblastoma (GBM) is one of the deadliest cancers. Treatment options are limited, and median patient survival is only several months. Translation of new therapies is hindered by a lack of GBM models that fully recapitulate disease heterogeneity. Here, we characterize two human GBM models (U87-luc2, U251-RedFLuc). In vitro, both cell lines express similar levels of luciferase and show comparable sensitivity to temozolomide and lapatinib exposure. In vivo, however, the two GBM models recapitulate different aspects of the disease. U87-luc2 cells quickly grow into large, well-demarcated tumors; U251-RedFLuc cells form small, highly invasive tumors. Using a new method to assess GBM invasiveness based on detecting tumor-specific anti-luciferase staining in brain slices, we found that U251-RedFLuc cells are more invasive than U87-luc2 cells. Lastly, we determined expression levels of ABC transporters in both models. Our findings indicate that U87-luc2 and U251-RedFLuc GBM models recapitulate different aspects of GBM heterogeneity that need to be considered in preclinical research.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA
| | - Louis T Rodgers
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA
| | - Richard J Kryscio
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, USA
- Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, USA
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy University of Kentucky, Lexington, KY, USA.
- Drug Discovery, Delivery and Translational Therapeutics Track, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, USA.
| |
Collapse
|
16
|
Todo T, Ito H, Ino Y, Ohtsu H, Ota Y, Shibahara J, Tanaka M. Intratumoral oncolytic herpes virus G47∆ for residual or recurrent glioblastoma: a phase 2 trial. Nat Med 2022; 28:1630-1639. [PMID: 35864254 PMCID: PMC9388376 DOI: 10.1038/s41591-022-01897-x] [Citation(s) in RCA: 254] [Impact Index Per Article: 84.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 06/09/2022] [Indexed: 12/23/2022]
Abstract
This investigator-initiated, phase 2, single-arm trial primarily assessed the efficacy of G47∆, a triple-mutated, third-generation oncolytic herpes simplex virus type 1, in 19 adult patients with residual or recurrent, supratentorial glioblastoma after radiation therapy and temozolomide (UMIN-CTR Clinical Trial Registry UMIN000015995). G47Δ was administered intratumorally and repeatedly for up to six doses. The primary endpoint of 1-yr survival rate after G47∆ initiation was 84.2% (95% confidence interval, 60.4-96.6; 16 of 19). The prespecified endpoint was met and the trial was terminated early. Regarding secondary endpoints, the median overall survival was 20.2 (16.8-23.6) months after G47∆ initiation and 28.8 (20.1-37.5) months from the initial surgery. The most common G47∆-related adverse event was fever (17 of 19) followed by vomiting, nausea, lymphocytopenia and leukopenia. On magnetic resonance imaging, enlargement of and contrast-enhancement clearing within the target lesion repeatedly occurred after each G47∆ administration, which was characteristic to this therapy. Thus, the best overall response in 2 yr was partial response in one patient and stable disease in 18 patients. Biopsies revealed increasing numbers of tumor-infiltrating CD4+/CD8+ lymphocytes and persistent low numbers of Foxp3+ cells. This study showed a survival benefit and good safety profile, which led to the approval of G47∆ as the first oncolytic virus product in Japan.
Collapse
Affiliation(s)
- Tomoki Todo
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, and Department of Surgical Neuro-Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Hirotaka Ito
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, and Department of Surgical Neuro-Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yasushi Ino
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, and Department of Surgical Neuro-Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Ohtsu
- Department of Data Science, National Center for Global Health and Medicine in Japan, Tokyo, Japan
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University, Tokyo, Japan
| | - Yasunori Ota
- Department of Pathology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Junji Shibahara
- Department of Pathology, Kyorin University School of Medicine, Tokyo, Japan
| | - Minoru Tanaka
- Division of Innovative Cancer Therapy, Advanced Clinical Research Center, and Department of Surgical Neuro-Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Caraway CA, Gaitsch H, Wicks EE, Kalluri A, Kunadi N, Tyler BM. Polymeric Nanoparticles in Brain Cancer Therapy: A Review of Current Approaches. Polymers (Basel) 2022; 14:2963. [PMID: 35890738 PMCID: PMC9322801 DOI: 10.3390/polym14142963] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
Translation of novel therapies for brain cancer into clinical practice is of the utmost importance as primary brain tumors are responsible for more than 200,000 deaths worldwide each year. While many research efforts have been aimed at improving survival rates over the years, prognosis for patients with glioblastoma and other primary brain tumors remains poor. Safely delivering chemotherapeutic drugs and other anti-cancer compounds across the blood-brain barrier and directly to tumor cells is perhaps the greatest challenge in treating brain cancer. Polymeric nanoparticles (NPs) are powerful, highly tunable carrier systems that may be able to overcome those obstacles. Several studies have shown appropriately-constructed polymeric NPs cross the blood-brain barrier, increase drug bioavailability, reduce systemic toxicity, and selectively target central nervous system cancer cells. While no studies relating to their use in treating brain cancer are in clinical trials, there is mounting preclinical evidence that polymeric NPs could be beneficial for brain tumor therapy. This review includes a variety of polymeric NPs and how their associated composition, surface modifications, and method of delivery impact their capacity to improve brain tumor therapy.
Collapse
Affiliation(s)
- Chad A. Caraway
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Hallie Gaitsch
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- NIH-Oxford-Cambridge Scholars Program, Wellcome—MRC Cambridge Stem Cell Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 1TN, UK
| | - Elizabeth E. Wicks
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
- University of Mississippi School of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Anita Kalluri
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Navya Kunadi
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| | - Betty M. Tyler
- Hunterian Neurosurgical Research Laboratory, Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; (C.A.C.); (H.G.); (E.E.W.); (A.K.); (N.K.)
| |
Collapse
|
18
|
Liu B, Zhang G, Cui S, Du G. Upregulation of KIF11 in TP53 Mutant Glioma Promotes Tumor Stemness and Drug Resistance. Cell Mol Neurobiol 2022; 42:1477-1485. [PMID: 33491154 PMCID: PMC11421707 DOI: 10.1007/s10571-020-01038-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/31/2020] [Indexed: 12/21/2022]
Abstract
Glioma is the most common type of primary brain malignancy with high morbidity and mortality, but little is known about its pathological mechanisms. Kinesin family member 11 (KIF11) is a key driver of malignancy in glioblastoma, a grade IV glioma, but its involvement in glioma chemoresistance remains to be determined. We accessed the TCGA open datasets, collected glioma tumor tissue samples, and analyzed the expression of KIF11 in glioma patients. Meanwhile, the correlation between KIF11 and survival outcomes was determined by the Kaplan-Meier analysis. The role of KIF11 in glioma tumor cell function was assessed in an in vitro knockdown and overexpressing system. Here, we found that KIF11 was upregulated in glioma tumors and negatively correlated with overall survival outcomes via analyzing the open datasets. KIF11 was negatively correlated with TP53 expression. Furthermore, KIF11 promoted the stemness in glioma cells, accompanied by increased cell proliferation and chemoresistance. Mechanistically, we found that KIF11 promoted cell cycle progression via upregulating cyclin expression.
Collapse
Affiliation(s)
- Bin Liu
- Department of Neurosurgery Six, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061000, Hebei, China.
| | - Gang Zhang
- Department of Neurosurgery Six, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Shukun Cui
- Department of Neurosurgery Six, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Guoliang Du
- Department of Neurosurgery Six, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061000, Hebei, China
| |
Collapse
|
19
|
Integration of synthetic and natural derivatives revives the therapeutic potential of temozolomide against glioma- an in vitro and in vivo perspective. Life Sci 2022; 301:120609. [DOI: 10.1016/j.lfs.2022.120609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/24/2022]
|
20
|
Pająk B. Looking for the Holy Grail—Drug Candidates for Glioblastoma Multiforme Chemotherapy. Biomedicines 2022; 10:biomedicines10051001. [PMID: 35625738 PMCID: PMC9138518 DOI: 10.3390/biomedicines10051001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 02/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the deadliest and the most heterogeneous brain cancer. The median survival time of GBM patients is approximately 8 to 15 months after initial diagnosis. GBM development is determined by numerous signaling pathways and is considered one of the most challenging and complicated-to-treat cancer types. Standard GBM therapy consist of surgery followed by radiotherapy or chemotherapy, and combined treatment. Current standard of care (SOC) does not offer a significant chance for GBM patients to combat cancer, and the selection of available drugs is limited. For almost 20 years, there has been only one drug, Temozolomide (TMZ), approved as a first-line GBM treatment. Due to the limited efficacy of TMZ and the high rate of resistant patients, the implementation of new chemotherapeutics is highly desired. However, due to the unique properties of GBM, many challenges still need to be overcome before reaching a ‘breakthrough’. This review article describes the most recent compounds introduced into clinical trials as drug candidates for GBM chemotherapy.
Collapse
Affiliation(s)
- Beata Pająk
- Independent Laboratory of Genetics and Molecular Biology, Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland
| |
Collapse
|
21
|
Xiang J, Ma L, Gu Z, Jin H, Zhai H, Tong J, Liang T, Li J, Ren Q, Liu Q. A Boronated Derivative of Temozolomide Showing Enhanced Efficacy in Boron Neutron Capture Therapy of Glioblastoma. Cells 2022; 11:cells11071173. [PMID: 35406737 PMCID: PMC8998031 DOI: 10.3390/cells11071173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/21/2022] [Accepted: 03/24/2022] [Indexed: 02/04/2023] Open
Abstract
There is an incontestable need for improved treatment modality for glioblastoma due to its extraordinary resistance to traditional chemoradiation therapy. Boron neutron capture therapy (BNCT) may play a role in the future. We designed and synthesized a 10B-boronated derivative of temozolomide, TMZB. BNCT was carried out with a total neutron radiation fluence of 2.4 ± 0.3 × 1011 n/cm2. The effects of TMZB in BNCT were measured with a clonogenic cell survival assay in vitro and PET/CT imaging in vivo. Then, 10B-boronated phenylalanine (BPA) was tested in parallel with TMZB for comparison. The IC50 of TMZB for the cytotoxicity of clonogenic cells in HS683 was 0.208 mM, which is comparable to the IC50 of temozolomide at 0.213 mM. In BNCT treatment, 0.243 mM TMZB caused 91.2% ± 6.4% of clonogenic cell death, while 0.239 mM BPA eliminated 63.7% ± 6.3% of clonogenic cells. TMZB had a tumor-to-normal brain ratio of 2.9 ± 1.1 and a tumor-to-blood ratio of 3.8 ± 0.2 in a mouse glioblastoma model. BNCT with TMZB in this model caused 58.2% tumor shrinkage at 31 days after neutron irradiation, while the number for BPA was 35.2%. Therefore, by combining the effects of chemotherapy from temozolomide and radiotherapy with heavy charged particles from BNCT, TMZB-based BNCT exhibited promising potential for therapeutic applications in glioblastoma treatment.
Collapse
Affiliation(s)
- Jing Xiang
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China;
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
| | - Lin Ma
- Department of Stomatology, General Hospital, Shenzhen University, Shenzhen 518055, China;
| | - Zheng Gu
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
| | - Hongjun Jin
- Guangdong Provincial Key Lab of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China;
| | - Hongbin Zhai
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
| | - Jianfei Tong
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Tianjiao Liang
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Juan Li
- Institute of High Energy Physics, Chinese Academy of Sciences (CAS), Beijing 100049, China; (J.T.); (T.L.); (J.L.)
- Spallation Neutron Source Science Center, Dongguan 523803, China
| | - Qiushi Ren
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China;
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
- Correspondence: (Q.R.); (Q.L.); Tel.: +86-0755-26038837 (Q.R. & Q.L.)
| | - Qi Liu
- Shenzhen Bay Laboratory, Institute of Biomedical Engineering, Shenzhen 518132, China;
- Institute of Biomedical Engineering, Peking University Shenzhen Graduate School, Shenzhen 518055, China;
- Correspondence: (Q.R.); (Q.L.); Tel.: +86-0755-26038837 (Q.R. & Q.L.)
| |
Collapse
|
22
|
Griffin CP, Paul CL, Alexander KL, Walker MM, Hondermarck H, Lynam J. Postmortem brain donations vs premortem surgical resections for glioblastoma research: viewing the matter as a whole. Neurooncol Adv 2022; 4:vdab168. [PMID: 35047819 PMCID: PMC8760897 DOI: 10.1093/noajnl/vdab168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
There have been limited improvements in diagnosis, treatment, and outcomes of primary brain cancers, including glioblastoma, over the past 10 years. This is largely attributable to persistent deficits in understanding brain tumor biology and pathogenesis due to a lack of high-quality biological research specimens. Traditional, premortem, surgical biopsy samples do not allow full characterization of the spatial and temporal heterogeneity of glioblastoma, nor capture end-stage disease to allow full evaluation of the evolutionary and mutational processes that lead to treatment resistance and recurrence. Furthermore, the necessity of ensuring sufficient viable tissue is available for histopathological diagnosis, while minimizing surgically induced functional deficit, leaves minimal tissue for research purposes and results in formalin fixation of most surgical specimens. Postmortem brain donation programs are rapidly gaining support due to their unique ability to address the limitations associated with surgical tissue sampling. Collecting, processing, and preserving tissue samples intended solely for research provides both a spatial and temporal view of tumor heterogeneity as well as the opportunity to fully characterize end-stage disease from histological and molecular standpoints. This review explores the limitations of traditional sample collection and the opportunities afforded by postmortem brain donations for future neurobiological cancer research.
Collapse
Affiliation(s)
- Cassandra P Griffin
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Cancer Biobank: NSW Regional Biospecimen and Research Services, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Cancer Research Alliance, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Christine L Paul
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Cancer Research Alliance, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Priority Research Centre Cancer Research, Innovation and Translation, University of Newcastle, New South Wales, Australia
- Priority Research Centre Health Behaviour, University of Newcastle, New South Wales, Australia
| | - Kimberley L Alexander
- Neurosurgery Department, Chris O’Brien Lifehouse, Camperdown, New South Wales, Australia
- Brainstorm Brain Cancer Research, Brain and Mind Centre, The University of Sydney, New South Wales, Australia
- Neuropathology Department, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Marjorie M Walker
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Cancer Research Alliance, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Hubert Hondermarck
- Hunter Cancer Research Alliance, University of Newcastle, Newcastle, New South Wales, Australia
- Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, New South Wales, Australia
| | - James Lynam
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Cancer Research Alliance, University of Newcastle, Newcastle, New South Wales, Australia
- Department of Medical Oncology, Calvary Mater, Newcastle, New South Wales, Australia
| |
Collapse
|
23
|
Zhang C, Wu J, Liu W, Zheng X, Zhang W, Lee CS, Wang P. A novel hypocrellin-based assembly for sonodynamic therapy against glioblastoma. J Mater Chem B 2021; 10:57-63. [PMID: 34842264 DOI: 10.1039/d1tb01886h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The non-invasive treatment of glioblastoma (GBM) is of great significance and can greatly reduce the complications of craniotomy. Sonodynamic therapy (SDT) is an emerging tumor therapeutic strategy that overcomes some fatal flaws of photodynamic therapy (PDT). Different from PDT, SDT has deep tissue penetration and can be applied in the non-invasive treatment of deep-seated tumors. However, effective sonosensitizers that can be used for SDT of GBM are still very rare. Herein, we have prepared a suitable assembly based on a hypocrellin derivative (CTHB) with good biocompatibility. Excitedly, the hypocrellin-based assembly (CTHB NPs) can effectively produce reactive oxygen species under ultrasound stimulation. The inherent fluorescence and photoacoustic imaging characteristics of the CTHB NPs are conducive to the precise positioning of the tumors. It has been proved both in subcutaneous and in intracranial tumor models that CTHB NPs can be used as an effective sonosensitizer to inhibit tumor growth under ultrasound irradiation. This hypocrellin-based assembly has a good clinical prospect in the non-invasive treatment of GBM.
Collapse
Affiliation(s)
- Chuangli Zhang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Jiasheng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China. .,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.
| | - Wenjun Zhang
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, People's Republic of China
| | - Chun-Sing Lee
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, People's Republic of China
| | - Pengfei Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China. .,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
24
|
Shabani M, Javanshir HT, Bereimipour A, Sadrabadi AE, Jalili A, Nayernia K. Contradictory Effect of Notch1 and Notch2 on Phosphatase and Tensin Homolog and its Influence on Glioblastoma Angiogenesis. Galen Med J 2021; 10:e2091. [PMID: 36643842 PMCID: PMC9829453 DOI: 10.31661/gmj.v10i0.2091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/22/2021] [Accepted: 05/28/2021] [Indexed: 01/18/2023] Open
Abstract
Many genes induce angiogenesis in tumors, and among them, Notch family genes have received particular attention due to their extensive network of connections with other genes active in this function. Suppression of angiogenic signaling has been studied in various cancers, confirming Notch's fundamental and extensive role. According to studies, four Notch genes work independently with many genes such as vascular endothelial growth factor, phosphatase and tensin homolog, Phosphoinositide 3-kinase/Akt, and matrix metalloproteinases, and so many other genes, as well as proteins (such as hypoxia-inducible factor-1 alpha) significantly affect tumor angiogenesis. Notch1 regular activity in a healthy person causes angiogenesis in body tissues, controlled by normal Notch2 activity. However, in many cases of glioblastoma, whether on patients or tumor xenografts or in vivo models, a mutation in one of these two essential genes or at least one of the genes and proteins that affected by them can cause better angiogenesis in hypoxic conditions and lead to become an invasive tumor. In this review, we examined the contrasting activity of Notch1 and Notch2 and the signaling cascade that each generates in the angiogenesis of glioblastoma, the most invasive cancer of the central nervous system.
Collapse
Affiliation(s)
- Mostafa Shabani
- Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hamid Taghvaei Javanshir
- Medical Genomics Research Center, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ahmad Bereimipour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Young Researchers and Elite Club, Tehran Medical Sciences Islamic Azad University, Tehran, Iran
| | - Amin Ebrahimi Sadrabadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Arsalan Jalili
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Karim Nayernia
- International Center for Personalized Medicine, Düsseldorf, Germany
- Karim Nayernia, International Center for Personalized Medicine, Düsseldorf, Germany. Telephone Number: +4921144773490 Email Address:
| |
Collapse
|
25
|
Yoo JY, Yeh M, Wang YY, Oh C, Zhao ZM, Kaur B, Lee TJ. MicroRNA-138 Increases Chemo-Sensitivity of Glioblastoma through Downregulation of Survivin. Biomedicines 2021; 9:biomedicines9070780. [PMID: 34356844 PMCID: PMC8301402 DOI: 10.3390/biomedicines9070780] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/28/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma (GBM) is one of the most deadly cancers and poorly responses to chemotherapies, such as temozolomide (TMZ). Dysregulation of intrinsic signaling pathways in cancer cells are often resulted by dysregulated tumor suppressive microRNAs (miRNAs). Previously, we found miR-138 as one of tumor suppressive miRNAs that were significantly down-regulated in GBM. In this study, we demonstrated that ectopic over-expression of miR-138 sensitizes GBM cells to the treatment of TMZ and increased apoptotic cell death. Mechanistically, miR-138 directly repressed the expression of Survivin, an anti-apoptotic protein, to enhance caspase-induced apoptosis upon TMZ treatment. Using an intracranial GBM xenograft mice model, we also showed that combination of miR-138 with TMZ increases survival rates of the mice compared to the control mice treated with TMZ alone. This study provides strong preclinical evidence of the therapeutic benefit from restoration of miR-138 to sensitize the GBM tumor to conventional chemotherapy.
Collapse
Affiliation(s)
- Ji-Young Yoo
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.-Y.Y.); (M.Y.); (B.K.)
| | - Margaret Yeh
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.-Y.Y.); (M.Y.); (B.K.)
| | - Yin-Ying Wang
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (Y.-Y.W.); (Z.-M.Z.)
| | - Christina Oh
- Department of Biosciences, Rice University, Houston, TX 77005, USA;
| | - Zhong-Ming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (Y.-Y.W.); (Z.-M.Z.)
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Balveen Kaur
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.-Y.Y.); (M.Y.); (B.K.)
| | - Tae-Jin Lee
- Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (J.-Y.Y.); (M.Y.); (B.K.)
- Correspondence:
| |
Collapse
|
26
|
Bruns J, Zustiak SP. Hydrogel-Based Spheroid Models of Glioblastoma for Drug Screening Applications. MISSOURI MEDICINE 2021; 118:346-351. [PMID: 34373670 PMCID: PMC8343644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor, with median patient survival of 12-15 months even after treatment. To facilitate basic research as well as treatment development, bioengineered GBM models that adequately recapitulate aspects of the in vivo tumor microenvironment are greatly needed. Multicellular spheroids are a well-accepted model in tumor biology as well as drug screening because they recapitulate many of the solid tumor characteristics, such as hypoxic core and cell-cell communication. There are multiple approaches for growing GBM cells into tumor spheroids - non-adherent plastic dishes, hanging drop, bioreactors, and hydrogels, amongst others. Suspension spheroid models offer ease of growth, uniformity, and overall lower cost, but neglect the cell-matrix interactions, while hydrogel-based spheroids capture cell-matrix interactions and allow co-cultures with stromal cells. In this review, we summarize various approaches to fabricate GBM spheroid models as well as GBM spheroid characteristics and chemotherapeutic responsiveness as a function of hydrogel matrix encapsulation and properties, in order to advance therapies.
Collapse
Affiliation(s)
| | - Silviya Petrova Zustiak
- Program of Biomedical Engineering and the Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri
| |
Collapse
|
27
|
Dang TT, Morales JC. Loss of CENP-I Impairs Homologous Recombination and Sensitizes Cells to PARP1 Inhibition. Cancers (Basel) 2021; 13:3202. [PMID: 34206916 PMCID: PMC8267748 DOI: 10.3390/cancers13133202] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
Centromere Protein I (CENP-I) is a member of the CENP-H/I/K complex. CENP-H/I/K is a major component of the inner kinetochore and aids in ensuring proper chromosomal segregation during mitosis. In addition to this chromosomal segregation function, CENP-I also plays a role in DNA double-strand break (DSB) repair. Loss of CENP-I leads to increased endogenous 53BP1 foci and R-loop formation, while reducing cellular survival after ionizing radiation and Niraparib, a PARP1 small molecule inhibitor, exposures. Cells lacking CENP-I display delayed 53BP1 foci regression, an indication that DSB repair is impaired. Additionally, loss of CENP-I impairs the homologous recombination DSB repair pathway, while having no effect on the non-homologous end-joining pathway. Interestingly, we find that RNaseH1 expression restores HR capacity in CENP-I deficient cells. Importantly, CENP-I expression is elevated in glioma tissue as compared to normal brain tissue. This elevated expression also correlates with poor overall patient survival. These data highlight the multi-functional role CENP-I plays in maintaining genetic, as well as chromosomal, stability and tumor survival.
Collapse
Affiliation(s)
| | - Julio C. Morales
- Stephenson Cancer Center, Department of Neurosurgery, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA;
| |
Collapse
|
28
|
Kirstein A, Schilling D, Combs SE, Schmid TE. Lomeguatrib Increases the Radiosensitivity of MGMT Unmethylated Human Glioblastoma Multiforme Cell Lines. Int J Mol Sci 2021; 22:ijms22136781. [PMID: 34202589 PMCID: PMC8268804 DOI: 10.3390/ijms22136781] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Treatment resistance of glioblastoma multiforme to chemo- and radiotherapy remains a challenge yet to overcome. In particular, the O6-methylguanine-DNA-methyltransferase (MGMT) promoter unmethylated patients have only little benefit from chemotherapy treatment using temozolomide since MGMT counteracts its therapeutic efficacy. Therefore, new treatment options in radiotherapy need to be developed to inhibit MGMT and increase radiotherapy response. Methods: Lomeguatrib, a highly specific MGMT inhibitor, was used to inactivate MGMT protein in vitro. Radiosensitivity of established human glioblastoma multiforme cell lines in combination with lomeguatrib was investigated using the clonogenic survival assay. Inhibition of MGMT was analyzed using Western Blot. Cell cycle distribution and apoptosis were investigated to determine the effects of lomeguatrib alone as well as in combination with ionizing radiation. Results: Lomeguatrib significantly decreased MGMT protein and reduced radiation-induced G2/M arrest. A radiosensitizing effect of lomeguatrib was observed when administered at 1 µM and increased radioresistance at 20 µM. Conclusion: Low concentrations of lomeguatrib elicit radiosensitization, while high concentrations mediate a radioprotective effect.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Daniela Schilling
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany; (A.K.); (D.S.); (S.E.C.)
- Department of Radiation Oncology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-3187-43040
| |
Collapse
|
29
|
Luo J, Li P. Human pluripotent stem cell-derived brain organoids as in vitro models for studying neural disorders and cancer. Cell Biosci 2021; 11:99. [PMID: 34049587 PMCID: PMC8161602 DOI: 10.1186/s13578-021-00617-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
The sheer complexities of brain and resource limitation of human brain tissue greatly hamper our understanding of the brain disorders and cancers. Recently developed three-dimensional (3D) brain organoids (BOs) are self-organized and spontaneously differentiated from human pluripotent stem cells (hPSCs) in vitro, which exhibit similar features with cell type diversity, structural organization, and functional connectivity as the developing human brain. Based on these characteristics, hPSC-derived BOs (hPDBOs) provide new opportunities to recapitulate the complicated processes during brain development, neurodegenerative disorders, and brain cancers in vitro. In this review, we will provide an overview of existing BO models and summarize the applications of this technology in modeling the neural disorders and cancers. Furthermore, we will discuss the challenges associated with their use as in vitro models for disease modeling and the potential future direction.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
30
|
Brennan PM, Borchert R, Coulter C, Critchley GR, Hall B, Holliman D, Phang I, Jefferies SJ, Keni S, Lee L, Liaquat I, Marcus HJ, Thomson S, Thorne L, Vintu M, Wiggins AN, Jenkinson MD, Erridge S. Second surgery for progressive glioblastoma: a multi-centre questionnaire and cohort-based review of clinical decision-making and patient outcomes in current practice. J Neurooncol 2021; 153:99-107. [PMID: 33791952 PMCID: PMC8131335 DOI: 10.1007/s11060-021-03748-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE Glioblastoma prognosis is poor. Treatment options are limited at progression. Surgery may benefit, but no quality guidelines exist to inform patient selection. We sought to describe variations in surgical management at progression, highlight where further evidence is needed, and build towards a consensus strategy. METHODS Current practice in selection of patients with progressive GBM for second surgery was surveyed online amongst specialists in the UK and Europe. We complemented this with an assessment of practice in a retrospective cohort study from six United Kingdom neurosurgical units. We used descriptive statistics to analyse the data. RESULTS 234 questionnaire responses were received. Maintaining or improving patient quality of life was key to decision making, with variation as to whether patient age, performance status or intended extent of resection was relevant. MGMT methylation status was not important. Half considered no minimum time after first surgery. 288 patients were reported in the cohort analysis. Median time to second surgery from first surgery 390 days. Median overall survival 815 days, with no association between time to second surgery and time to death (p = 0.874). CONCLUSIONS This is the most wide-ranging examination of contemporaneous practice in management of GBM progression. Without evidence-based guidelines, the variation is unsurprising. We propose consensus guidelines for consideration, to reduce heterogeneity in decision making, support data collection and analysis of factors influencing outcomes, and to inform clinical trials to establish whether second surgery improves patient outcomes, or simply selects to patients already performing well.
Collapse
Affiliation(s)
- P M Brennan
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh BioQuarter, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| | - R Borchert
- Addenbrookes University Hospital, Cambridge, UK
| | - C Coulter
- Royal Victoria Hospital, Newcastle, UK
| | - G R Critchley
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - B Hall
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | | | - I Phang
- Lancashire teaching Hospitals, Preston, UK
| | | | - S Keni
- University of Edinburgh medical School, Edinburgh, UK
| | - L Lee
- University of Edinburgh medical School, Edinburgh, UK
| | - I Liaquat
- Department of Clinical Neuroscience, NHS Lothian, Edinburgh, UK
| | - H J Marcus
- National Hospital for Neurology and Neurosurgery, Queen Square, London, UK
| | | | - L Thorne
- University College London Hospitals, London, UK
| | - M Vintu
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | - A N Wiggins
- Department of Clinical Neuroscience, NHS Lothian, Edinburgh, UK
| | - M D Jenkinson
- Institute of Translational Medicine, University of Liverpool, Liverpool, UK
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Liverpool, UK
| | - S Erridge
- Department of Clinical Neuroscience, NHS Lothian, Edinburgh, UK
| |
Collapse
|
31
|
Arora P, Gudelsky G, Desai PB. Gender-based differences in brain and plasma pharmacokinetics of letrozole in sprague-dawley rats: Application of physiologically-based pharmacokinetic modeling to gain quantitative insights. PLoS One 2021; 16:e0248579. [PMID: 33798227 PMCID: PMC8018653 DOI: 10.1371/journal.pone.0248579] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/01/2021] [Indexed: 01/04/2023] Open
Abstract
Based on the discovery that the estrogen synthase aromatase (CYP19A1) is abundantly expressed in high- grade gliomas, the aromatase inhibitor, letrozole is being investigated in pre-clinical models as a novel agent against this malignancy. Here, we investigated the systemic and brain pharmacokinetics of letrozole following single and steady state dosing in both male and female Sprague-Dawley rats. Furthermore, we employed physiologically-based pharmacokinetic (PBPK) modeling to gain quantitative insights into the blood-brain barrier penetration of this drug. Letrozole (4 mg/kg) was administered intraperitoneally daily for 5 days (for males) and 11 days (for females) and intracerebral microdialysis was performed for brain extracellular fluid (ECF) collection simultaneously with venous blood sampling. Drug levels were measured using HPLC and non-compartmental analysis was conducted employing WinNonlin®. Simcyp animal simulator was used for conducting bottom-up PBPK approach incorporating the specified multi-compartment brain model. Overall, marked gender-specific differences in the systemic and brain pharmacokinetics of letrozole were observed. Letrozole clearance was much slower in female rats resulting in markedly higher plasma and brain drug concentrations. At steady state, the plasma AUC 0-24 was 103.0 and 24.8 μg*h/ml and brain ECF AUC 0-12 was 24.0 and 4.8 μg*h/ml in female and male rats, respectively. The PBPK model simulated brain concentration profiles were in close agreement with the observed profiles. While gender-specific differences in letrozole PK are not observed in the clinical setting, these findings will guide the dose optimization during pre-clinical investigations of this compound. The PBPK model will serve as an important clinical translational tool.
Collapse
Affiliation(s)
- Priyanka Arora
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Gary Gudelsky
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Pankaj B Desai
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio, United States of America
| |
Collapse
|
32
|
Anticancer Mechanism of Curcumin on Human Glioblastoma. Nutrients 2021; 13:nu13030950. [PMID: 33809462 PMCID: PMC7998496 DOI: 10.3390/nu13030950] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most malignant brain tumor and accounts for most adult brain tumors. Current available treatment options for GBM are multimodal, which include surgical resection, radiation, and chemotherapy. Despite the significant advances in diagnostic and therapeutic approaches, GBM remains largely resistant to treatment, with a poor median survival rate between 12 and 18 months. With increasing drug resistance, the introduction of phytochemicals into current GBM treatment has become a potential strategy to combat GBM. Phytochemicals possess multifarious bioactivities with multitarget sites and comparatively marginal toxicity. Among them, curcumin is the most studied compound described as a potential anticancer agent due to its multi-targeted signaling/molecular pathways properties. Curcumin possesses the ability to modulate the core pathways involved in GBM cell proliferation, apoptosis, cell cycle arrest, autophagy, paraptosis, oxidative stress, and tumor cell motility. This review discusses curcumin’s anticancer mechanism through modulation of Rb, p53, MAPK, P13K/Akt, JAK/STAT, Shh, and NF-κB pathways, which are commonly involved and dysregulated in preclinical and clinical GBM models. In addition, limitation issues such as bioavailability, pharmacokinetics perspectives strategies, and clinical trials were discussed.
Collapse
|
33
|
Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [ 18F]DASA-23. Mol Imaging Biol 2021; 22:124-133. [PMID: 30989436 DOI: 10.1007/s11307-019-01353-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET). PROCEDURE U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6 days (n = 6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake. RESULTS Significant interactions were found between the treatment agent (n = 12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p = 0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p < 0. 0001) followed by irinotecan (p = 0. 0012), erlotinib (p = 0. 02), and 5-fluorouracil (p = 0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r = 0.44, p = 0.15). CONCLUSIONS These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.
Collapse
|
34
|
Paranthaman S, Goravinahalli Shivananjegowda M, Mahadev M, Moin A, Hagalavadi Nanjappa S, Nanjaiyah ND, Chidambaram SB, Gowda DV. Nanodelivery Systems Targeting Epidermal Growth Factor Receptors for Glioma Management. Pharmaceutics 2020; 12:pharmaceutics12121198. [PMID: 33321953 PMCID: PMC7763629 DOI: 10.3390/pharmaceutics12121198] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/17/2020] [Accepted: 10/18/2020] [Indexed: 02/06/2023] Open
Abstract
A paradigm shift in treating the most aggressive and malignant form of glioma is continuously evolving; however, these strategies do not provide a better life and survival index. Currently, neurosurgical debulking, radiotherapy, and chemotherapy are the treatment options available for glioma, but these are non-specific in action. Patients invariably develop resistance to these therapies, leading to recurrence and death. Receptor Tyrosine Kinases (RTKs) are among the most common cell surface proteins in glioma and play a significant role in malignant progression; thus, these are currently being explored as therapeutic targets. RTKs belong to the family of cell surface receptors that are activated by ligands which in turn activates two major downstream signaling pathways via Rapidly Accelerating Sarcoma/mitogen activated protein kinase/extracellular-signal-regulated kinase (Ras/MAPK/ERK) and phosphatidylinositol 3-kinase/a serine/threonine protein kinase/mammalian target of rapamycin (PI3K/AKT/mTOR). These pathways are critically involved in regulating cell proliferation, invasion, metabolism, autophagy, and apoptosis. Dysregulation in these pathways results in uncontrolled glioma cell proliferation, invasion, angiogenesis, and cancer progression. Thus, RTK pathways are considered a potential target in glioma management. This review summarizes the possible risk factors involved in the growth of glioblastoma (GBM). The role of RTKs inhibitors (TKIs) and the intracellular signaling pathways involved, small molecules under clinical trials, and the updates were discussed. We have also compiled information on the outcomes from the various endothelial growth factor receptor (EGFR)-TKIs-based nanoformulations from the preclinical and clinical points of view. Aided by an extensive literature search, we propose the challenges and potential opportunities for future research on EGFR-TKIs-based nanodelivery systems.
Collapse
Affiliation(s)
- Sathishbabu Paranthaman
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (S.P.); (M.G.S.); (M.M.)
| | | | - Manohar Mahadev
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (S.P.); (M.G.S.); (M.M.)
| | - Afrasim Moin
- Department of Pharmaceutics, Hail University, Hail PO BOX 2440, Saudi Arabia;
| | | | | | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India;
| | - Devegowda Vishakante Gowda
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, India; (S.P.); (M.G.S.); (M.M.)
- Correspondence: ; Tel.: +91-9663162455
| |
Collapse
|
35
|
Tan B, Huang L, Wu Y, Liao J. Advances and trends of hydrogel therapy platform in localized tumor treatment: A review. J Biomed Mater Res A 2020; 109:404-425. [PMID: 32681742 DOI: 10.1002/jbm.a.37062] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 02/04/2023]
Abstract
Due to limitations of treatment and the stubbornness of infiltrative tumor cells, the outcome of conventional antitumor treatment is often compromised by a variety of factors, including severe side effects, unexpected recurrence, and massive tissue loss during the treatment. Hydrogel-based therapy is becoming a promising option of cancer treatment, because of its controllability, biocompatibility, high drug loading, prolonged drug release, and specific stimuli-sensitivity. Hydrogel-based therapy has good malleability and can reach some areas that cannot be easily touched by surgeons. Furthermore, hydrogel can be used not only as a carrier for tumor treatment agents, but also as a scaffold for tissue repair. In this review, we presented the latest researches in hydrogel applications of localized tumor therapy and highlighted the recent progress of hydrogel-based therapy in preventing postoperative tumor recurrence and improving tissue repair, thus proposing a new trend of hydrogel-based technology in localized tumor therapy. And this review aims to provide a novel reference and inspire thoughts for a more accurate and individualized cancer treatment.
Collapse
Affiliation(s)
- Bowen Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lingxiao Huang
- Department of Basic Research, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yongzhi Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
O’Connor KP, Palejwala AH, Milton CK, Lu VM, Glenn CA, Sughrue ME, Conner AK. Laser Interstitial Thermal Therapy Case Series: Choosing the Correct Number of Fibers Depending on Lesion Size. Oper Neurosurg (Hagerstown) 2020; 20:18-23. [DOI: 10.1093/ons/opaa264] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/18/2020] [Indexed: 12/21/2022] Open
Abstract
Abstract
BACKGROUND
Laser interstitial thermal therapy (LITT) is being used for the treatment of recurrent glioblastoma multiforme (GBM). Lesions can be treated using 1 or multiple LITT fibers depending on the preference of surgeons. Usually, more fibers are needed for coverage of larger tumors.
OBJECTIVE
To investigate and analyze how tumor size affected the number of LITT fibers used.
METHODS
This is a retrospective review of patients undergoing treatment of recurrent GBM. Patients were treated with up to 4 LITT fibers for adequate tumor coverage. Patient demographics, tumor characteristics, length of stay, complications, and biopsy results were recorded.
RESULTS
A total of 43 cases were treated using LITT, and of these cases, 31 consisted of contiguous lesions. We used more fibers to treat larger tumor volumes. On average, for each 5 cc of tumor volume, a fiber was added for proper coverage (P = .554). Complications and length of stay were similar across the groups (P = .378, P = .941).
CONCLUSION
LITT can be used for the treatment of recurrent GBM. For each 5 cc of tumor volume, a LITT fiber can be added to the treatment plan.
Collapse
Affiliation(s)
- Kyle P O’Connor
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Ali H Palejwala
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Camille K Milton
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Victor M Lu
- Department of Neurosurgery, Mayo Clinic, Rochester, Minnesota
| | - Chad A Glenn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Michael E Sughrue
- Department of Neurosurgery, Prince of Wales Hospital, Randwick, Australia
| | - Andrew K Conner
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
37
|
Soni N, Ora M, Mohindra N, Menda Y, Bathla G. Diagnostic Performance of PET and Perfusion-Weighted Imaging in Differentiating Tumor Recurrence or Progression from Radiation Necrosis in Posttreatment Gliomas: A Review of Literature. AJNR Am J Neuroradiol 2020; 41:1550-1557. [PMID: 32855194 DOI: 10.3174/ajnr.a6685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/29/2020] [Indexed: 01/22/2023]
Abstract
Tumor resection followed by chemoradiation remains the current criterion standard treatment for high-grade gliomas. Regardless of aggressive treatment, tumor recurrence and radiation necrosis are 2 different outcomes. Differentiation of tumor recurrence from radiation necrosis remains a critical problem in these patients because of considerable overlap in clinical and imaging presentations. Contrast-enhanced MR imaging is the universal imaging technique for diagnosis, treatment evaluation, and detection of recurrence of high-grade gliomas. PWI and PET with novel radiotracers have an evolving role for monitoring treatment response in high-grade gliomas. In the literature, there is no clear consensus on the superiority of either technique or their complementary information. This review aims to elucidate the diagnostic performance of individual and combined use of functional (PWI) and metabolic (PET) imaging modalities to distinguish recurrence from posttreatment changes in gliomas.
Collapse
Affiliation(s)
- N Soni
- Department of Radiology (N.S., Y.M., G.B.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - M Ora
- Department of Radiodiagnosis (M.O., N.M.), Sanjay Gandhi Post Graduate Institute of Medical Sciences, Institute of Nuclear Medicine, Lucknow, India
| | - N Mohindra
- Department of Radiodiagnosis (M.O., N.M.), Sanjay Gandhi Post Graduate Institute of Medical Sciences, Institute of Nuclear Medicine, Lucknow, India
| | - Y Menda
- Department of Radiology (N.S., Y.M., G.B.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - G Bathla
- Department of Radiology (N.S., Y.M., G.B.), University of Iowa Hospitals and Clinics, Iowa City, Iowa
| |
Collapse
|
38
|
The effects of point-source electroporation on the blood-brain barrier and brain vasculature in rats: An MRI and histology study. Bioelectrochemistry 2020; 134:107521. [DOI: 10.1016/j.bioelechem.2020.107521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/31/2022]
|
39
|
Lin EY, Chen YS, Li YS, Chen SR, Lee CH, Huang MH, Chuang HM, Harn HJ, Yang HH, Lin SZ, Tai DF, Chiou TW. Liposome Consolidated with Cyclodextrin Provides Prolonged Drug Retention Resulting in Increased Drug Bioavailability in Brain. Int J Mol Sci 2020; 21:ijms21124408. [PMID: 32575820 PMCID: PMC7352271 DOI: 10.3390/ijms21124408] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/11/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Although butylidenephthalide (BP) is an efficient anticancer drug, its poor bioavailability renders it ineffective for treating drug-resistant brain tumors. However, this problem is overcome through the use of noninvasive delivery systems, including intranasal administration. Herein, the bioavailability, drug stability, and encapsulation efficiency (EE, up to 95%) of BP were improved by using cyclodextrin-encapsulated BP in liposomal formulations (CDD1). The physical properties and EE of the CDD1 system were investigated via dynamic light scattering, transmission electron microscopy, UV–Vis spectroscopy, and nuclear magnetic resonance spectroscopy. The cytotoxicity was examined via MTT assay, and the cellular uptake was observed using fluorescence microscopy. The CDD1 system persisted for over 8 h in tumor cells, which was a considerable improvement in the retention of the BP-containing cyclodextrin or the BP-containing liposomes, thereby indicating a higher BP content in CDD1. Nanoscale CDD1 formulations were administered intranasally to nude mice that had been intracranially implanted with temozolomide-resistant glioblastoma multiforme cells, resulting in increased median survival time. Liquid chromatography–mass spectrometry revealed that drug biodistribution via intranasal delivery increased the accumulation of BP 10-fold compared to oral delivery methods. Therefore, BP/cyclodextrin/liposomal formulations have potential clinical applications for treating drug-resistant brain tumors.
Collapse
Affiliation(s)
- En-Yi Lin
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan; (E.-Y.L.); (Y.-S.L.); (S.-R.C.); (C.-H.L.)
- Department of Chemistry, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
| | - Yu-Shuan Chen
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan;
| | - Yuan-Sheng Li
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan; (E.-Y.L.); (Y.-S.L.); (S.-R.C.); (C.-H.L.)
| | - Syuan-Rong Chen
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan; (E.-Y.L.); (Y.-S.L.); (S.-R.C.); (C.-H.L.)
| | - Chia-Hung Lee
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan; (E.-Y.L.); (Y.-S.L.); (S.-R.C.); (C.-H.L.)
| | - Mao-Hsuan Huang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
- Department of Stem Cell Applied Technology, Gwo Xi Stem Cell Applied Technology, Hsinchu 30261, Taiwan
| | - Hong-Meng Chuang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
- Laboratory of Translational Medicine Office, Development Center for Biotechnology, Taipei 115, Taiwan
| | - Horng-Jyh Harn
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
- Department of Pathology, Hualien Tzu Chi Hospital, Tzu Chi University, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Hsueh-Hui Yang
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan;
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (Y.-S.C.); (M.-H.H.); (H.-M.C.); (H.-J.H.); (S.-Z.L.)
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Dar-Fu Tai
- Department of Chemistry, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan
- Correspondence: (D.-F.T.); (T.-W.C.); Tel.: +886-3-890-3579 (D.-F.T.); +886-3-890-3638 (T.-W.C.); Fax: +886-3-890-0162 (D.-F.T.); +886-3-890-0398 (T.-W.C.)
| | - Tzyy-Wen Chiou
- Department of Life Science, National Dong Hwa University, No. 1, Sec. 2, Da Hsueh Rd., Shou-Feng, Hualien 974301, Taiwan; (E.-Y.L.); (Y.-S.L.); (S.-R.C.); (C.-H.L.)
- Correspondence: (D.-F.T.); (T.-W.C.); Tel.: +886-3-890-3579 (D.-F.T.); +886-3-890-3638 (T.-W.C.); Fax: +886-3-890-0162 (D.-F.T.); +886-3-890-0398 (T.-W.C.)
| |
Collapse
|
40
|
Zhang C, Wu J, Liu W, Zheng X, Zhang W, Lee CS, Wang P. Hypocrellin-Based Multifunctional Phototheranostic Agent for NIR-Triggered Targeted Chemo/Photodynamic/Photothermal Synergistic Therapy against Glioblastoma. ACS APPLIED BIO MATERIALS 2020; 3:3817-3826. [PMID: 35025252 DOI: 10.1021/acsabm.0c00386] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A huge challenge exists in the diagnosis and treatment of malignant glioblastoma (GBM) due to the presence of the blood-brain barrier (BBB). Herein, a multifunctional phototheranostic agent is designed on the basis of an octadecane-modified temozolomide (TMZ-C18) for chemotherapy, a dicysteamine-modified hypocrellin derivative (DCHB) as a natural-origin photosensitizer with a singlet oxygen (1O2) quantum yield of 0.51, and a cyclic peptide (cRGD) as a targeting unit against glioblastoma. Co-encapsulated DCHB and TMZ-C18 assembly with cRGD decoration, referred to as DTRGD NPs, shows a wide absorption at the NIR region peaked at 703 nm, an NIR emission peak at 720 nm, good photostability, high photothermal conversion efficiency (33%), and effective degradation of TMZ-C18. More importantly, DTRGD NPs can efficiently break through the blood-brain barrier and enrich in the orthotopic glioblastoma. The treatment of subcutaneous U87MG tumor beard mice demonstrates that DTRGD NPs present remarkable anticancer efficiency and the targeted chemo/photodynamic/photothermal synergistic therapy can be achieved with almost no toxicity. This multifunctional phototheranostic agent shows great potential for the diagnosis and treatment of glioblastoma.
Collapse
Affiliation(s)
- Chuangli Zhang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jiasheng Wu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Wenjun Zhang
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Chun-Sing Lee
- Center of Super-Diamond and Advanced Films (COSDAF) & Department of Materials Science and Engineering, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, P. R. China
| | - Pengfei Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
41
|
Involvement of POLA2 in Double Strand Break Repair and Genotoxic Stress. Int J Mol Sci 2020; 21:ijms21124245. [PMID: 32549188 PMCID: PMC7352189 DOI: 10.3390/ijms21124245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/04/2023] Open
Abstract
Cellular survival is dependent on the efficient replication and transmission of genomic information. DNA damage can be introduced into the genome by several different methods, one being the act of DNA replication. Replication is a potent source of DNA damage and genomic instability, especially through the formation of DNA double strand breaks (DSBs). DNA polymerase alpha is responsible for replication initiation. One subunit of the DNA polymerase alpha replication machinery is POLA2. Given the connection between replication and genomic instability, we decided to examine the role of POLA2 in DSB repair, as little is known about this topic. We found that loss of POLA2 leads to an increase in spontaneous DSB formation. Loss of POLA2 also slows DSB repair kinetics after treatment with etoposide and inhibits both of the major double strand break repair pathways: non-homologous end-joining and homologous recombination. In addition, loss of POLA2 leads to increased sensitivity to ionizing radiation and PARP1 inhibition. Lastly, POLA2 expression is elevated in glioblastoma multiforme tumors and correlates with poor overall patient survival. These data demonstrate a role for POLA2 in DSB repair and resistance to genotoxic stress.
Collapse
|
42
|
Tran TAT, Kim YH, Duong THO, Jung S, Kim IY, Moon KS, Jang WY, Lee HJ, Lee JJ, Jung TY. Peptide Vaccine Combined Adjuvants Modulate Anti-tumor Effects of Radiation in Glioblastoma Mouse Model. Front Immunol 2020; 11:1165. [PMID: 32733437 PMCID: PMC7358653 DOI: 10.3389/fimmu.2020.01165] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 05/12/2020] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma, the most common aggressive cancer, has a poor prognosis. Among the current standard treatment strategies, radiation therapy is the most commonly recommended. However, it is often unsuccessful at completely eliminating the cancer from the brain. A combination of radiation with other treatment methods should therefore be considered. It has been reported that radiotherapy in combination with immunotherapy might show a synergistic effect; however, this still needs to be investigated. In the current study, a “branched multipeptide and peptide adjuvants [such as pan DR epitope (PADRE) and polyinosinic-polycytidylic acid—stabilized with polylysine and carboxymethylcellulose—(poly-ICLC)],” namely vaccine and anti-PD1, were used as components of immunotherapy to assist in the anti-tumor effects of radiotherapy against glioblastomas. With regard to experimental design, immunological characterization of GL261 cells was performed and the effects of radiation on this cell line were also evaluated. An intracranial GL261 mouse glioma model was established, and therapeutic effects were observed based on tumor size and survival time. The distribution of effector immune cells in the spleen, based on cytotoxic T lymphocyte (CTL) and natural killer (NK) cell function, was determined. The pro-inflammatory and anti-inflammatory cytokine production from re-stimulated splenocytes and single tumor cells were also evaluated. As GL261 cells demonstrated both immunological characteristics and radiation sensitivity, they were found to be promising candidates for testing this combination treatment. Combinatorial treatment with radiation, vaccine, and anti-PD1 prolonged mouse survival by delaying tumor growth. Although this combination treatment led to an increase in the functional activity of both CTLs and NK cells, as evidenced by the increased percentage of these cells in the spleen, there was a greater shift toward CTL rather than NK cell activity. Moreover, the released cytokines from re-stimulated splenocytes and single tumor cells also showed a shift toward the pro-inflammatory response. This study suggests that immunotherapy comprising a branched multipeptide plus PADRE, poly-ICLC, and anti-PD1 could potentially enhance the anti-tumor effects of radiotherapy in a glioblastoma mouse model.
Collapse
Affiliation(s)
- Thi-Anh-Thuy Tran
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Young-Hee Kim
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Thi-Hoang-Oanh Duong
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Shin Jung
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - In-Young Kim
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Kyung-Sub Moon
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Woo-Youl Jang
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Hyun-Ju Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| | - Tae-Young Jung
- Brain Tumor Research Laboratory, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea.,Department of Neurosurgery, Chonnam National University Medical School and Hwasun Hospital, Chonnam National University, Hwasun, South Korea
| |
Collapse
|
43
|
Sun W, Zhang W, Yu J, Lu Z, Yu J. Inhibition of Nrf2 might enhance the anti-tumor effect of temozolomide in glioma cells via inhibition of Ras/Raf/MEK signaling pathway. Int J Neurosci 2020; 131:975-983. [PMID: 32378973 DOI: 10.1080/00207454.2020.1766458] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Glioblastoma (GBM) is the most common aggressive primary cancer occurring in the brain tissue. GBM accounts 16% of primary brain tumors and half of gliomas. Additionally, the incidence of GBM is increases with aging, and reaches the peak at the age of 75 to 84 years. The survival of patients with GBM remains at a low level, only less than 5% patients diagnosed with GBM survive for 5 years. Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM. TMZ combined with radiation therapy has been shown to prolong the overall survival (OS) to 14.6 months compared with 12.1 months for radiation therapy alone. NF-E2-related factor 2 (Nrf2) is a transcription factor that contains seven functional domains. The binding of Keap1 to Nrf2 is a central regulator of the cellular defense mechanism against environmental stresses. METHODS First, Nrf2 overexpression and inhibition models were constructed in U251 cells using transfection. The percentage of viable cells was detected using the MTT assay. Then, the expression of the HO-1 regulator was detected using qPCR, and the concentrations of oxidative stress related factors were detected using ELISAs. The levels of proteins related to oxidative stress and the Ras/Raf/MEK signaling pathway was detected using western blotting analysis. RESULTS We initially established Nrf2 inhibition and activation cell models in U251 cells and found that the inhibition of Nrf2 expression decreased the mRNA and protein levels of the anti-oxidative enzymes, as well as the secretion of these enzymes into the cellular microenvironment. These effects might be mediated by the inhibition of Ras/Raf/MEK signaling pathway, leading to the inhibition of cellular proliferation. CONCLUSIONS Inhibition of Nrf2 expression might enhance the effect of TMZ on the treatment of GBM and might be a new therapeutic strategy.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurosurgery, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Weihua Zhang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianyong Yu
- Department of Pharmacy, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Zhihui Lu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianhua Yu
- Department of Oncology, Jiangxi Provincial People's Hospital, Nanchang, China
| |
Collapse
|
44
|
Kim MS, Lim J, Shin HS, Cho KG. Re-Irradiation and Its Contribution to Good Prognosis in Recurrent Glioblastoma Patients. Brain Tumor Res Treat 2020; 8:29-35. [PMID: 32390351 PMCID: PMC7221471 DOI: 10.14791/btrt.2020.8.e10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/11/2020] [Accepted: 03/17/2020] [Indexed: 01/02/2023] Open
Abstract
Background Radiation therapy, one of the strongest anti-cancer treatments, is already performed to treat primary glioblastoma; however, the effect of repeated radiation therapy for recurrent tumors has not been fully explored. The aim of this study was to determine the efficacy of re-irradiation in treating recurrent glioblastoma. Methods The study included 36 patients with recurrent glioblastoma treated with repeated radiation therapy between 2002 and 2016. Stereotactic radiosurgery (SRS) and hypo-fractionated stereotactic radiotherapy (HSRT) were performed in these patients. Results Fourteen patients received SRS with a median dose of 25 Gy (range, 20–32 Gy) in 1–5 fractions. Twenty-two patients received HSRT with a median dose of 40 Gy (range, 31.5–52 Gy) in 6–20 fractions. There were six treatment-related grade 3 adverse events. Survival analysis showed that re-irradiation significantly prolonged overall survival (OS) and progression-free survival (PFS). The median OS and one-year OS rate after re-irradiation were 17.2 months and 60.4%, respectively. The median PFS and 6-month PFS rate after re-irradiation were 4.4 months and 41.9%, respectively. Of the 36 patients, three survived without any progression in their condition. Conclusion Re-irradiation for recurrent glioblastoma showed favorable outcomes. Radiation dose and fractionation should be carefully considered to minimize radiation necrosis.
Collapse
Affiliation(s)
- Mi Sun Kim
- Department of Radiation Oncology, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam, Korea
| | - Jaejoon Lim
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam, Korea
| | - Hyun Soo Shin
- Department of Radiation Oncology, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam, Korea
| | - Kyung Gi Cho
- Department of Neurosurgery, Bundang CHA Medical Center, CHA University College of Medicine, Seongnam, Korea.
| |
Collapse
|
45
|
Kirstein A, Schmid TE, Combs SE. The Role of miRNA for the Treatment of MGMT Unmethylated Glioblastoma Multiforme. Cancers (Basel) 2020; 12:cancers12051099. [PMID: 32354046 PMCID: PMC7281574 DOI: 10.3390/cancers12051099] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most common high-grade intracranial tumor in adults. It is characterized by uncontrolled proliferation, diffuse infiltration due to high invasive and migratory capacities, as well as intense resistance to chemo- and radiotherapy. With a five-year survival of less than 3% and an average survival rate of 12 months after diagnosis, GBM has become a focus of current research to urgently develop new therapeutic approaches in order to prolong survival of GBM patients. The methylation status of the promoter region of the O6-methylguanine–DNA methyltransferase (MGMT) is nowadays routinely analyzed since a methylated promoter region is beneficial for an effective response to temozolomide-based chemotherapy. Furthermore, several miRNAs were identified regulating MGMT expression, apart from promoter methylation, by degrading MGMT mRNA before protein translation. These miRNAs could be a promising innovative treatment approach to enhance Temozolomide (TMZ) sensitivity in MGMT unmethylated patients and to increase progression-free survival as well as long-term survival. In this review, the relevant miRNAs are systematically reviewed.
Collapse
Affiliation(s)
- Anna Kirstein
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Thomas E. Schmid
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Stephanie E. Combs
- Institute of Radiation Medicine (IRM), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany
- Correspondence: ; Tel.: +49-89-4140-4501
| |
Collapse
|
46
|
Hua TNM, Oh J, Kim S, Antonio JM, Vo VTA, Om J, Choi JW, Kim JY, Jung CW, Park MJ, Jeong Y. Peroxisome proliferator-activated receptor gamma as a theragnostic target for mesenchymal-type glioblastoma patients. Exp Mol Med 2020; 52:629-642. [PMID: 32280134 PMCID: PMC7210935 DOI: 10.1038/s12276-020-0413-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 01/18/2023] Open
Abstract
Glioblastomas (GBMs) are characterized by four subtypes, proneural (PN), neural, classical, and mesenchymal (MES) GBMs, and they all have distinct activated signaling pathways. Among the subtypes, PN and MES GBMs show mutually exclusive genetic signatures, and the MES phenotype is, in general, believed to be associated with more aggressive features of GBM: tumor recurrence and drug resistance. Therefore, targeting MES GBMs would improve the overall prognosis of patients with fatal tumors. In this study, we propose peroxisome proliferator-activated receptor gamma (PPARγ) as a potential diagnostic and prognostic biomarker as well as therapeutic target for MES GBM; we used multiple approaches to assess PPARγ, including biostatistics analysis and assessment of preclinical studies. First, we found that PPARγ was exclusively expressed in MES glioblastoma stem cells (GSCs), and ligand activation of endogenous PPARγ suppressed cell growth and stemness in MES GSCs. Further in vivo studies involving orthotopic and heterotopic xenograft mouse models confirmed the therapeutic efficacy of targeting PPARγ; compared to control mice, those that received ligand treatment exhibited longer survival as well as decreased tumor burden. Mechanistically, PPARγ activation suppressed proneural-mesenchymal transition (PMT) by inhibiting the STAT3 signaling pathway. Biostatistical analysis using The Cancer Genomics Atlas (TCGA, n = 206) and REMBRANDT (n = 329) revealed that PPARγ upregulation is linked to poor overall survival and disease-free survival of GBM patients. Analysis was performed on prospective (n = 2) and retrospective (n = 6) GBM patient tissues, and we finally confirmed that PPARγ expression was distinctly upregulated in MES GBM. Collectively, this study provides insight into PPARγ as a potential therapeutic target for patients with MES GBM.
Collapse
Affiliation(s)
- Tuyen N M Hua
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Jiwoong Oh
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Sohyun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jayson M Antonio
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Vu T A Vo
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea
| | - Jiyeon Om
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
| | - Jong-Whan Choi
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea
| | - Jeong-Yub Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Chan-Woong Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Myung-Jin Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea.
| | - Yangsik Jeong
- Departments of Biochemistry, Yonsei University, Wonju, Republic of Korea.
- Departments of Global Medical Science, Yonsei University, Wonju, Republic of Korea.
- Departments of Mitohormesis Research Center, Yonsei University, Wonju, Republic of Korea.
- Institutes of Lifestyle Medicine, Yonsei University, Wonju, Republic of Korea.
- Departments of Mitochondrial Medicine, Yonsei University, Wonju, Republic of Korea.
- Departments of Nuclear Receptor Research Consortium, Wonju College of Medicine, Yonsei University, Wonju, Gangwon-Do, 26426, Republic of Korea.
| |
Collapse
|
47
|
Silvia N, Dai G. CEREBRAL ORGANOIDS AS A MODEL FOR GLIOBLASTOMA MULTIFORME. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 13:152-159. [PMID: 32355905 DOI: 10.1016/j.cobme.2020.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly lethal and elusive cancer. While many in vitro and in vivo models have been developed to recapitulate the factors that contribute to its invasive behavior, they suffer from drawbacks related to genetic variability, expense and scope. Technologies utilizing human pluripotent stem cells can now generate organoids which can recapitulate the relative complexity the cytoarchitecture and microenvironment of human brain tissue. In conjunction with protocols which effectively induce GBM tumors within these "cerebral organoids", such approaches represent an unprecedented model to investigate GBM invasion and its effect on the brain ECM. This review focuses on methods of brain organoid development, protocols for inducing GBM, the relevant findings on invasion and microenvironmental changes, and discusses their limitations and potential future direction.
Collapse
Affiliation(s)
- Nathaniel Silvia
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
48
|
Kalasauskas D, Sorokin M, Sprang B, Elmasri A, Viehweg S, Salinas G, Opitz L, Rave-Fraenk M, Schulz-Schaeffer W, Kantelhardt SR, Giese A, Buzdin A, Kim EL. Diversity of Clinically Relevant Outcomes Resulting from Hypofractionated Radiation in Human Glioma Stem Cells Mirrors Distinct Patterns of Transcriptomic Changes. Cancers (Basel) 2020; 12:cancers12030570. [PMID: 32121554 PMCID: PMC7139840 DOI: 10.3390/cancers12030570] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/12/2020] [Accepted: 02/22/2020] [Indexed: 12/17/2022] Open
Abstract
Hypofractionated radiotherapy is the mainstay of the current treatment for glioblastoma. However, the efficacy of radiotherapy is hindered by the high degree of radioresistance associated with glioma stem cells comprising a heterogeneous compartment of cell lineages differing in their phenotypic characteristics, molecular signatures, and biological responses to external signals. Reconstruction of radiation responses in glioma stem cells is necessary for understanding the biological and molecular determinants of glioblastoma radioresistance. To date, there is a paucity of information on the longitudinal outcomes of hypofractionated radiation in glioma stem cells. This study addresses long-term outcomes of hypofractionated radiation in human glioma stem cells by using a combinatorial approach integrating parallel assessments of the tumor-propagating capacity, stemness-associated properties, and array-based profiling of gene expression. The study reveals a broad spectrum of changes in the tumor-propagating capacity of glioma stem cells after radiation and finds association with proliferative changes at the onset of differentiation. Evidence is provided that parallel transcriptomic patterns and a cumulative impact of pathways involved in the regulation of apoptosis, neural differentiation, and cell proliferation underly similarities in tumorigenicity changes after radiation.
Collapse
Affiliation(s)
- Darius Kalasauskas
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (D.K.); (B.S.); (A.E.); (S.V.)
- Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany;
| | - Maxim Sorokin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (M.S.); (A.B.)
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Omicsway Corp., Walnut, CA 91789, USA
| | - Bettina Sprang
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (D.K.); (B.S.); (A.E.); (S.V.)
| | - Alhassan Elmasri
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (D.K.); (B.S.); (A.E.); (S.V.)
| | - Sina Viehweg
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (D.K.); (B.S.); (A.E.); (S.V.)
| | - Gabriela Salinas
- NGS Integrative Genomics Core Unit (NIG), Institute for Human Genetics, University Medical Centre, 37077 Göttingen, Germany; (G.S.); (L.O.)
| | - Lennart Opitz
- NGS Integrative Genomics Core Unit (NIG), Institute for Human Genetics, University Medical Centre, 37077 Göttingen, Germany; (G.S.); (L.O.)
| | - Margret Rave-Fraenk
- Department of Radiotherapy and Radiooncology, University Medical Centre, 37077 Göttingen, Germany;
| | | | - Sven Reiner Kantelhardt
- Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany;
| | - Alf Giese
- OrthoCentrum Hamburg, Department of Tumor Spinal Surgery, 20149 Hamburg, Germany;
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; (M.S.); (A.B.)
- I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Oncobox ltd., 121205 Moscow, Russia
- Moscow Institute of Physics and Technology (National Research University), 141700 Moscow, Russia
| | - Ella L. Kim
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Centre, 55131 Mainz, Germany; (D.K.); (B.S.); (A.E.); (S.V.)
- Correspondence:
| |
Collapse
|
49
|
Ius T, Pignotti F, Della Pepa GM, La Rocca G, Somma T, Isola M, Battistella C, Gaudino S, Polano M, Dal Bo M, Bagatto D, Pegolo E, Chiesa S, Arcicasa M, Olivi A, Skrap M, Sabatino G. A Novel Comprehensive Clinical Stratification Model to Refine Prognosis of Glioblastoma Patients Undergoing Surgical Resection. Cancers (Basel) 2020; 12:E386. [PMID: 32046132 PMCID: PMC7072471 DOI: 10.3390/cancers12020386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/29/2020] [Accepted: 02/05/2020] [Indexed: 12/14/2022] Open
Abstract
Despite recent discoveries in genetics and molecular fields, glioblastoma (GBM) prognosis still remains unfavorable with less than 10% of patients alive 5 years after diagnosis. Numerous studies have focused on the research of biological biomarkers to stratify GBM patients. We addressed this issue in our study by using clinical/molecular and image data, which is generally available to Neurosurgical Departments in order to create a prognostic score that can be useful to stratify GBM patients undergoing surgical resection. By using the random forest approach [CART analysis (classification and regression tree)] on Survival time data of 465 cases, we developed a new prediction score resulting in 10 groups based on extent of resection (EOR), age, tumor volumetric features, intraoperative protocols and tumor molecular classes. The resulting tree was trimmed according to similarities in the relative hazard ratios amongst groups, giving rise to a 5-group classification tree. These 5 groups were different in terms of overall survival (OS) (p < 0.000). The score performance in predicting death was defined by a Harrell's c-index of 0.79 (95% confidence interval [0.76-0.81]). The proposed score could be useful in a clinical setting to refine the prognosis of GBM patients after surgery and prior to postoperative treatment.
Collapse
Affiliation(s)
- Tamara Ius
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Fabrizio Pignotti
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
| | | | - Giuseppe La Rocca
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Università degli Studi di Napoli Federico II, 80131 Naples, Italy;
| | - Miriam Isola
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Claudio Battistella
- Department of Medicine, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy; (M.I.); (C.B.)
| | - Simona Gaudino
- Institute of radiology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy; (M.P.); (M.D.B.)
| | - Daniele Bagatto
- Neuroradiology Unit, Department of Diagnostic Imaging ASUIUD Udine, 33100 Udine, Italy;
| | - Enrico Pegolo
- Institute of Pathology, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Silvia Chiesa
- Radiation Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Mauro Arcicasa
- Department of Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy;
| | - Alessandro Olivi
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| | - Miran Skrap
- Neurosurgery Unit, Department of Neuroscience, Santa Maria della Misericordia University Hospital, 33100 Udine, Italy;
| | - Giovanni Sabatino
- Department of Neurosurgery, Mater Olbia Hospital, 07026 Olbia, Italy; (F.P.); (G.S.); (G.L.R.)
- Institute of Neurosurgery, Catholic University, 00168 Rome, Italy; (G.M.D.P.); (A.O.)
| |
Collapse
|
50
|
Inhibition of phosphatidylinositol 3-kinase by PX-866 suppresses temozolomide-induced autophagy and promotes apoptosis in glioblastoma cells. Mol Med 2019; 25:49. [PMID: 31726966 PMCID: PMC6854621 DOI: 10.1186/s10020-019-0116-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022] Open
Abstract
Background Temozolomide (TMZ) is the most commonly used chemotherapeutic agent used to treat glioblastoma (GBM), which causes significant DNA damage to highly proliferative cells. Our observations have added to accumulating evidence that TMZ induces stress-responsive cellular programs known to promote cell survival, including autophagy. As such, targeting these survival pathways may represent new vulnerabilities of GBM after treatment with TMZ. Methods Using the T98G human glioma cell line, we assessed the molecular signaling associated with TMZ treatment, the cellular consequences of using the pan-PI3K inhibitor PX-866, and performed clonogenic assays to determine the effect sequential treatment of TMZ and PX-866 had on colony formation. Additionally, we also use subcutaneous GBM patient derived xenograft (PDX) tumors to show relative LC3 protein expression and correlations between survival pathways and molecular markers which dictate clinical responsiveness to TMZ. Results Here, we report that TMZ can induce autophagic flux in T98G glioma cells. GBM patient-derived xenograft (PDX) tumors treated with TMZ also display an increase in the autophagosome marker LC3 II. Additionally, O6-methylguanine-DNA-methyltransferase (MGMT) expression correlates with PI3K/AKT activity, suggesting that patients with inherent resistance to TMZ (MGMT-high) would benefit from PI3K/AKT inhibitors in addition to TMZ. Accordingly, we have identified that the blood-brain barrier (BBB) penetrant pan-PI3K inhibitor, PX-866, is an early-stage inhibitor of autophagic flux, while maintaining its ability to inhibit PI3K/AKT signaling in glioma cells. Lastly, due to the induction of autophagic flux by TMZ, we provide evidence for sequential treatment of TMZ followed by PX-866, rather than combined co-treatment, as a means to shut down autophagy-induced survival in GBM cells and to enhance apoptosis. Conclusions The understanding of how TMZ induces survival pathways, such as autophagy, may offer new therapeutic vulnerabilities and opportunities to use sequential inhibition of alternate pro-survival pathways that regulate autophagy. As such, identification of additional ways to inhibit TMZ-induced autophagy could enhance the efficacy of TMZ.
Collapse
|