1
|
Liu L, Liao C, Ge S, Liu X, Dong J, Weng S, Huang G, Zhang Z, Jin Q, Wang J, Fan X, Zhang KN, Jiang T. Aggressive resection of non-contrast-enhanced tumor provides varying benefits to glioblastoma, IDH-wildtype patients based on different clinical characteristics. Cancer Lett 2025; 612:217471. [PMID: 39842498 DOI: 10.1016/j.canlet.2025.217471] [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: 11/07/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/24/2025]
Abstract
Supramaximal resection in glioblastoma, concerning non-contrast-enhancing (nCE) tumors, exhibited additional survival benefits. However, whether all patients can benefit from supramaximal resection of nCE tumors and the optimal resection target remains unclear, especially for the glioblastoma, IDH-wildtype under the new WHO CNS tumor classification. Clinical and surgical characteristics were collected from 155 patients with newly diagnosed glioblastoma, IDH-wildtype from the Chinese Glioma Genome Atlas, and a prospective cohort of 128 patients was enrolled for external validation. Recursive partitioning analysis was used to identify risk groups considering the effects of residual nCE tumor volume (RnTV) and clinical factors on overall survival (OS). Age, preoperative Karnofsky Performance Score (KPS), MGMT promoter status, and postoperative RnTV were independently associated with patient survival. Four risk groups with distinct prognoses were identified: Group 1 (median OS: 13.4 months), RnTV >43.27 ml; Group 2 (median OS: 17.8 months), RnTV ≤43.27 ml, KPS ≤90, and age ≥60; Group 3 (median OS: 22.3 months), RnTV 5.27-43.27 ml, age <60; Group 4 (median OS: 38.2 months) including 4a, KPS 100 and RnTV ≤43.27 ml; and 4b, KPS ≤90, age <60, and RnTV ≤5.27 ml. These results were retained regardless of MGMT promoter methylation status and validated in the external prospective validation cohort. Supramaximal nCE tumor resection enhances survival outcomes in glioblastoma, IDH-wildtype, but depending on clinical characteristics. In young symptomatic patients, supramaximal resection should be recommended with the RnTV ≤5.27 ml; in symptomless patients or elder patients, keeping the RnTV ≤43.27 is recommended to obtain the survival benefit from tumor resection surgery.
Collapse
Affiliation(s)
- Lingyu Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Chihyi Liao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Siqi Ge
- Department of Neuroepidemiology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xing Liu
- Department of Neuropathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiahan Dong
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Shimeng Weng
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guoshi Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Qiang Jin
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jiangwei Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Xing Fan
- Department of Neuroelectrophysiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Ke-Nan Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Ahmady F, Sharma A, Achuthan AA, Kannourakis G, Luwor RB. The Role of TIM-3 in Glioblastoma Progression. Cells 2025; 14:346. [PMID: 40072074 PMCID: PMC11899008 DOI: 10.3390/cells14050346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
Several immunoregulatory or immune checkpoint receptors including T cell immunoglobulin and mucin domain 3 (TIM-3) have been implicated in glioblastoma progression. Rigorous investigation over the last decade has elucidated TIM-3 as a key player in inhibiting immune cell activation and several key associated molecules have been identified both upstream and downstream that mediate immune cell dysfunction mechanistically. However, despite several reviews being published on other immune checkpoint molecules such as PD-1 and CTLA-4 in the glioblastoma setting, no such extensive review exists that specifically focuses on the role of TIM-3 in glioblastoma progression and immunosuppression. Here, we critically summarize the current literature regarding TIM-3 expression as a prognostic marker for glioblastoma, its expression profile on immune cells in glioblastoma patients and the exploration of anti-TIM-3 agents in glioblastoma pre-clinical models for potential clinical application.
Collapse
Affiliation(s)
- Farah Ahmady
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, 53127 Bonn, Germany;
- Department of Neurosurgery, University Hospital Bonn, 53127 Bonn, Germany
| | - Adrian A. Achuthan
- Department of Medicine, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3350, Australia;
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
| | - Rodney B. Luwor
- Fiona Elsey Cancer Research Institute, Ballarat, VIC 3350, Australia; (F.A.); (G.K.)
- Federation University, Ballarat, VIC 3350, Australia
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC 3350, Australia
- Huagene Institute, Kecheng Science and Technology Park, Pukou District, Nanjing 211806, China
| |
Collapse
|
3
|
Yu S, Wu J, Jing Y, Lin P, Lang L, Xiong Y, Chen W, Liu W, Sun C, Lu Y. Research trends in glioma chemoradiotherapy resistance: a bibliometric analysis (2003-2023). Front Oncol 2025; 15:1539937. [PMID: 39990688 PMCID: PMC11842341 DOI: 10.3389/fonc.2025.1539937] [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: 12/05/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
Background Glioma is the most aggressive primary malignant tumor of the central nervous system, characterized by high recurrence rates and resistance to chemoradiotherapy, making therapeutic resistance a major challenge in neuro-oncology. Recent research emphasizes the role of the tumor microenvironment (TME) and immune modulation in glioma progression and resistance. Despite these advances, a comprehensive bibliometric analysis of research trends in glioma chemoradiotherapy resistance over the past two decades is lacking. This study aims to systematically evaluate the research landscape, identify emerging hotspots, and provide guidance for future investigations. Methods Articles on glioma chemoradiotherapy resistance published between 2003 and 2023 were retrieved from the Web of Science Core Collection, resulting in 4,528 publications. Bibliometric tools, including VOSviewer, CiteSpace, and R packages such as bibliometrix and ggplot2, were used to analyze co-authorship networks, keyword evolution, and citation bursts to identify collaboration patterns, thematic developments, and influential contributions. Results Publication output increased significantly between 2013 and 2022, peaking at 650 articles in 2022. Over 1,000 institutions from 88 countries contributed to this research. The United States, Switzerland, and Germany showed the highest citation impact, while China led in publication volume but demonstrated relatively lower citation influence. The research focus has shifted from traditional topics such as the "MGMT gene" to emerging areas including the "tumor microenvironment," "immune infiltration," and "nanoparticles." The androgen receptor was identified as a promising but underexplored therapeutic target. Conclusions Research on glioma chemoradiotherapy resistance has seen substantial growth, with increasing emphasis on immune modulation, the tumor microenvironment, and novel therapeutic targets such as the androgen receptor. This study represents the first comprehensive bibliometric analysis of this field, providing a detailed overview of research trends and potential directions for future studies. The findings highlight the need for strengthened international collaboration and multidisciplinary approaches to address the challenges of therapeutic resistance in glioma.
Collapse
Affiliation(s)
- Shishi Yu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinya Wu
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuan Jing
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Lin
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Lang Lang
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifan Xiong
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wangzhong Chen
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenhua Liu
- Clinical Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Changpeng Sun
- The Editorial Department of the Journal of Southern Medical University, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuntao Lu
- Nanfang hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Sadeghipour A, Fattahi F, Madjd Z, Tajik F, Sedaghati F, Saeednejad Zanjani L. Increased nuclear expression of DNA damage inducible transcript 4 can serve as a potential prognostic biomarker in patients with gliomas: a study based on data mining and experimental tools. Discov Oncol 2025; 16:124. [PMID: 39915428 PMCID: PMC11802953 DOI: 10.1007/s12672-025-01865-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
INTRODUCTION DNA damage-inducible transcript 4 (DDIT4), induced under cellular stress conditions, has been implicated in malignancies due to its abnormal expression patterns. MATERIALS AND METHODS The expression of DDIT4 at the mRNA level and its potential role as a prognostic biomarker in gliomas were analyzed using the GEPIA tool. To validate these findings, DDIT4 protein expression levels and their prognostic significance were examined in tissue microarrays from glioma patients using immunohistochemistry in clinical samples. RESULTS Bioinformatics analysis revealed that DDIT4 overexpression in glioma tumors and its high mRNA expression levels are associated with poor outcomes, likely through mTOR signaling. At the protein level, positive nuclear DDIT4 expression was linked to higher histological grades and temozolomide treatment. Kaplan-Meier survival analysis demonstrated that positive nuclear DDIT4 expression is a significant predictor of poor disease-specific survival (DSS) and recurrence-free survival (RFS). Additionally, nuclear DDIT4 expression was identified as an independent prognostic factor for DSS. CONCLUSION Our findings suggest that nuclear DDIT4 expression may serve as a potential prognostic biomarker and therapeutic target in glioma patients. However, further studies with larger sample sizes and long-term follow-ups are needed to validate these observations and confirm their clinical relevance.
Collapse
Affiliation(s)
- Alireza Sadeghipour
- Department of Pathology, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Clinical Research Development Unit of Ayatollah-Khansari Hospital, Arak University of Medical Sciences, Arak, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Surgery, Irvine Medical Center, University of California, Orange, CA, USA
| | - Farnoosh Sedaghati
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
5
|
Lhotska H, Janeckova K, Cechova H, Macoun J, Aghova T, Lizcova L, Svobodova K, Hodanova L, Konecna D, Soukup J, Kramar F, Netuka D, Zemanova Z. Validating a clinically based MS-MLPA threshold through comparison with Sanger sequencing in glioblastoma patients. Clin Epigenetics 2025; 17:16. [PMID: 39881389 PMCID: PMC11776323 DOI: 10.1186/s13148-025-01822-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Glioblastoma is the commonest malignant brain tumor and has a very poor prognosis. Reduced expression of the MGMT gene (10q26.3), influenced primarily by the methylation of two differentially methylated regions (DMR1 and DMR2), is associated with a good response to temozolomide treatment. However, suitable methods for detecting the methylation of the MGMT gene promoter and setting appropriate cutoff values are debated. RESULTS A cohort of 108 patients with histologically and genetically defined glioblastoma was retrospectively examined with methylation-specific Sanger sequencing (sSeq) and methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) methods. The DMR2 region was methylated in 29% of samples, whereas DMR1 was methylated in 12% of samples. Methylation detected with the MS-MLPA method using probes MGMT_215, MGMT_190, and MGMT_124 from the ME012-A1 kit (located in DMR1 and DMR2) correlated with the methylation of the corresponding CpG dinucleotides detected with sSeq (p = 0.005 for probe MGMT_215; p < 0.001 for probe MGMT_190; p = 0.016 for probe MGMT_124). The threshold for methylation detection with the MS-MLPA method was calculated with a ROC curve analysis and principal components analysis of the data obtained with the MS-MLPA and sSeq methods, yielding a weighted value of 0.362. Thus, methylation of the MGMT gene promoter was confirmed in 36% of samples. These patients had statistically significantly better overall survival (p = 0.003). CONCLUSIONS Our results show that the threshold for methylation detection with the MS-MLPA method determined here is useful from a diagnostic perspective because it allows the stratification of patients who will benefit from specific treatment protocols, including temozolomide. Detailed analysis of the MGMT gene promoter enables the more-precise and personalized treatment of patients with glioblastoma.
Collapse
Affiliation(s)
- Halka Lhotska
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Karolina Janeckova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Hana Cechova
- Department of HLA, Institute of Hematology and Blood Transfusion, U Nemocnice 2094/1, 128 00, Prague, Czech Republic
| | - Jaromir Macoun
- The Clinical Trials and Research Department, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Tatiana Aghova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Libuse Lizcova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Karla Svobodova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Lucie Hodanova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic
| | - Dora Konecna
- Department of Neurosurgery and Neurooncology, 1st Faculty of Medicine of Charles University and Military University Hospital Prague, U Vojenske Nemocnice 1200, 169 02, Prague, Czech Republic
| | - Jiri Soukup
- Department of Pathology, 1st Faculty of Medicine of Charles University and Military University Hospital Prague, U Vojenske Nemocnice 1200, 169 02, Prague, Czech Republic
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine in Hradec Kralove and University Hospital Hradec Kralove, Sokolska 581, 500 05, Hradec Kralove, Czech Republic
| | - Filip Kramar
- Department of Neurosurgery and Neurooncology, 1st Faculty of Medicine of Charles University and Military University Hospital Prague, U Vojenske Nemocnice 1200, 169 02, Prague, Czech Republic
| | - David Netuka
- Department of Neurosurgery and Neurooncology, 1st Faculty of Medicine of Charles University and Military University Hospital Prague, U Vojenske Nemocnice 1200, 169 02, Prague, Czech Republic
| | - Zuzana Zemanova
- Center of Oncocytogenomics, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital and 1st Faculty of Medicine of Charles University in Prague, U Nemocnice 499/2, 128 00, Prague, Czech Republic.
| |
Collapse
|
6
|
Bruhn H, Tavelin B, Rosenlund L, Henriksson R. Do presenting symptoms predict treatment decisions and survival in glioblastoma? Real-world data from 1458 patients in the Swedish brain tumor registry. Neurooncol Pract 2024; 11:652-659. [PMID: 39279780 PMCID: PMC11398927 DOI: 10.1093/nop/npae036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Background Glioblastoma is the most common malignant brain tumor in adults. Non-invasive clinical parameters could play a crucial role in treatment planning and serve as predictors of patient survival. Our register-based real-life study aimed to investigate the prognostic value of presenting symptoms. Methods Data on presenting symptoms and survival, as well as known prognostic factors, were retrieved for all glioblastoma patients in Sweden registered in the Swedish Brain Tumor Registry between 2018 and 2021. The prognostic impact of different presenting symptoms was calculated using the Cox proportional hazard model. Results Data from 1458 adults with pathologically verified IDH wild-type glioblastoma were analyzed. Median survival time was 345 days. The 2-year survival rate was 21.5%. Registered presenting symptoms were focal neurological deficits, cognitive dysfunction, headache, epilepsy, signs of raised intracranial pressure, and cranial nerve symptoms, with some patients having multiple symptoms. Patients with initial cognitive dysfunction had significantly shorter survival than patients without; 265 days (245-285) vs. 409 days (365-453; P < .001). The reduced survival remained after Cox regression adjusting for known prognostic factors. Patients presenting with seizures and patients with headaches had significantly longer overall survival compared to patients without these symptoms, but the difference was not retained in multivariate analysis. Patients with cognitive deficits were less likely to have radical surgery and to receive extensive anti-neoplastic nonsurgical treatment. Conclusions This extensive real-life study reveals that initial cognitive impairment acts as an independent negative predictive factor for treatment decisions and adversely affects survival outcomes in glioblastoma patients.
Collapse
Affiliation(s)
- Helena Bruhn
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Björn Tavelin
- Clinical Research Unit, Cancercentrum, Region Vasterbotten, Umea University Hospital, Umea, Sweden
| | | | - Roger Henriksson
- Department of Radiation Sciences, Oncology, Umea University Hospital, Umea, Sweden
| |
Collapse
|
7
|
Azambuja JH, Yerneni SS, Maurer LM, Crentsil HE, Debom GN, Klei L, Smyers M, Sneiderman CT, Schwab KE, Acharya R, Wu YL, Ekambaram P, Hu D, Gough PJ, Bertin J, Melnick A, Kohanbash G, Bao R, Lucas PC, McAllister-Lucas LM. MALT1 protease inhibition restrains glioblastoma progression by reversing tumor-associated macrophage-dependent immunosuppression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.614808. [PMID: 39386586 PMCID: PMC11463364 DOI: 10.1101/2024.09.26.614808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
MALT1 protease is an intracellular signaling molecule that promotes tumor progression via cancer cell-intrinsic and cancer cell-extrinsic mechanisms. MALT1 has been mostly studied in lymphocytes, and little is known about its role in tumor-associated macrophages. Here, we show that MALT1 plays a key role in glioblastoma (GBM)-associated macrophages. Mechanistically, GBM tumor cells induce a MALT1-NF-κB signaling axis within macrophages, leading to macrophage migration and polarization toward an immunosuppressive phenotype. Inactivation of MALT1 protease promotes transcriptional reprogramming that reduces migration and restores a macrophage "M1-like" phenotype. Preclinical in vivo analysis shows that MALT1 inhibitor treatment results in increased immuno-reactivity of GBM-associated macrophages and reduced GBM tumor growth. Further, the addition of MALT1 inhibitor to temozolomide reduces immunosuppression in the tumor microenvironment, which may enhance the efficacy of this standard-of-care chemotherapeutic. Together, our findings suggest that MALT1 protease inhibition represents a promising macrophage-targeted immunotherapeutic strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Juliana Hofstätter Azambuja
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
| | - Saigopalakrishna S. Yerneni
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Chemical Engineering, Carnegie Mellon University; Pittsburgh, Pennsylvania
| | - Lisa M. Maurer
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Hannah E. Crentsil
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- Medical Scientist Training Program (MSTP), University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Gabriela N. Debom
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Linda Klei
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Mei Smyers
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Chaim T. Sneiderman
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Kristina E. Schwab
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh; Pittsburgh, Pennsylvania
| | | | - Yijen Lin Wu
- Rangos Research Center Animal Imaging Core, UPMC Children’s Hospital of Pittsburgh; Pittsburgh, Pennsylvania
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Dong Hu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- Department of Pathology, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Pete J. Gough
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline; King of Prussia, Pennsylvania
| | - John Bertin
- Pattern Recognition Receptor Discovery Performance Unit, Immuno-Inflammation Therapeutic Area, GlaxoSmithKline; King of Prussia, Pennsylvania
| | - Ari Melnick
- Division of Hematology and Oncology, Cornell University, New York, New York
| | - Gary Kohanbash
- Department of Neurological Surgery, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
| | - Riyue Bao
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh; Pittsburgh, Pennsylvania
| | - Peter C. Lucas
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Pathology, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Linda M. McAllister-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine; Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center; Pittsburgh, Pennsylvania
- Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
- Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
8
|
Huo X, Li H, Xing Y, Liu W, Chen P, Du F, Song L, Yu Z, Cao X, Tian J. Two decades of progress in glioma methylation research: the rise of temozolomide resistance and immunotherapy insights. Front Neurosci 2024; 18:1440756. [PMID: 39286478 PMCID: PMC11402815 DOI: 10.3389/fnins.2024.1440756] [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: 05/30/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Aims This study aims to systematically analyze the global trends in glioma methylation research using bibliometric methodologies. We focus on identifying the scholarly trajectory and key research interests, and we utilize these insights to predict future research directions within the epigenetic context of glioma. Methods We performed a comprehensive literature search of the Web of Science Core Collection (WoSCC) to identify articles related to glioma methylation published from January 1, 2004, to December 31, 2023. The analysis included full-text publications in the English language and excluded non-research publications. Analysis and visualization were performed using GraphPad Prism, CiteSpace, and VOSviewer software. Results The search identified 3,744 publications within the WoSCC database, including 3,124 original research articles and 620 review articles. The research output gradually increased from 2004 to 2007, followed by a significant increase after 2008, which peaked in 2022. A minor decline in publication output was noted during 2020-2021, potentially linked to the coronavirus disease 2019 pandemic. The United States and China were the leading contributors, collectively accounting for 57.85% of the total research output. The Helmholtz Association of Germany, the German Cancer Research Center (DKFZ), and the Ruprecht Karls University of Heidelberg were the most productive institutions. The Journal of Neuro-Oncology led in terms of publication volume, while Neuro-Oncology had the highest Impact Factor. The analysis of publishing authors revealed Michael Weller as the most prolific contributor. The co-citation network analysis identified David N. Louis's article as the most frequently cited. The keyword analysis revealed "temozolomide," "expression," "survival," and "DNA methylation" as the most prominent keywords, while "heterogeneity," "overall survival," and "tumor microenvironment" showed the strongest citation bursts. Conclusions The findings of this study illustrate the increasing scholarly interest in glioma methylation, with a notable increase in research output over the past two decades. This study provides a comprehensive overview of the research landscape, highlighting the importance of temozolomide, DNA methylation, and the tumor microenvironment in glioma research. Despite its limitations, this study offers valuable insights into the current research trends and potential future directions, particularly in the realm of immunotherapy and epigenetic editing techniques.
Collapse
Affiliation(s)
- Xianhao Huo
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Haoyuan Li
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Yixiang Xing
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Wenqing Liu
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Pengfei Chen
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Fang Du
- School of Information Engineering, Ningxia University, Yinchuan, China
- Collaborative Innovation Center for Ningxia Big Data and Artificial Intelligence Co-founded by Ningxia Municipality and Ministry of Education, Ningxia University, Yinchuan, China
| | - Lijuan Song
- School of Information Engineering, Ningxia University, Yinchuan, China
| | - Zhenhua Yu
- School of Information Engineering, Ningxia University, Yinchuan, China
| | - Xiangmei Cao
- Basic Medical School, Ningxia Medical University, Yinchuan, China
| | - Jihui Tian
- Department of Neurosurgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
9
|
Fazzari E, Azizad DJ, Yu K, Ge W, Li MX, Nano PR, Kan RL, Tum HA, Tse C, Bayley NA, Haka V, Cadet D, Perryman T, Soto JA, Wick B, Raleigh DR, Crouch EE, Patel KS, Liau LM, Deneen B, Nathanson DA, Bhaduri A. Glioblastoma Neurovascular Progenitor Orchestrates Tumor Cell Type Diversity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.24.604840. [PMID: 39091877 PMCID: PMC11291138 DOI: 10.1101/2024.07.24.604840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Glioblastoma (GBM) is the deadliest form of primary brain tumor with limited treatment options. Recent studies have profiled GBM tumor heterogeneity, revealing numerous axes of variation that explain the molecular and spatial features of the tumor. Here, we seek to bridge descriptive characterization of GBM cell type heterogeneity with the functional role of individual populations within the tumor. Our lens leverages a gene program-centric meta-atlas of published transcriptomic studies to identify commonalities between diverse tumors and cell types in order to decipher the mechanisms that drive them. This approach led to the discovery of a tumor-derived stem cell population with mixed vascular and neural stem cell features, termed a neurovascular progenitor (NVP). Following in situ validation and molecular characterization of NVP cells in GBM patient samples, we characterized their function in vivo. Genetic depletion of NVP cells resulted in altered tumor cell composition, fewer cycling cells, and extended survival, underscoring their critical functional role. Clonal analysis of primary patient tumors in a human organoid tumor transplantation system demonstrated that the NVP has dual potency, generating both neuronal and vascular tumor cells. Although NVP cells comprise a small fraction of the tumor, these clonal analyses demonstrated that they strongly contribute to the total number of cycling cells in the tumor and generate a defined subset of the whole tumor. This study represents a paradigm by which cell type-specific interrogation of tumor populations can be used to study functional heterogeneity and therapeutically targetable vulnerabilities of GBM.
Collapse
Affiliation(s)
- Elisa Fazzari
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Daria J Azizad
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Kwanha Yu
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Weihong Ge
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Matthew X Li
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Patricia R Nano
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Ryan L Kan
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Hong A Tum
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Christopher Tse
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas A Bayley
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Vjola Haka
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dimitri Cadet
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Travis Perryman
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Jose A Soto
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Brittney Wick
- Genomics Institute, University of California Santa Cruz, Santa Cruz, CA, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, USA
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
| | - Elizabeth E Crouch
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, USA
| | - Kunal S Patel
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - David A Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Pridham KJ, Hutchings KR, Beck P, Liu M, Xu E, Saechin E, Bui V, Patel C, Solis J, Huang L, Tegge A, Kelly DF, Sheng Z. Selective regulation of chemosensitivity in glioblastoma by phosphatidylinositol 3-kinase beta. iScience 2024; 27:109921. [PMID: 38812542 PMCID: PMC11133927 DOI: 10.1016/j.isci.2024.109921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 05/31/2024] Open
Abstract
Resistance to chemotherapies such as temozolomide is a major hurdle to effectively treat therapy-resistant glioblastoma. This challenge arises from the activation of phosphatidylinositol 3-kinase (PI3K), which makes it an appealing therapeutic target. However, non-selectively blocking PI3K kinases PI3Kα/β/δ/γ has yielded undesired clinical outcomes. It is, therefore, imperative to investigate individual kinases in glioblastoma's chemosensitivity. Here, we report that PI3K kinases were unequally expressed in glioblastoma, with levels of PI3Kβ being the highest. Patients deficient of O6-methylguanine-DNA-methyltransferase (MGMT) and expressing elevated levels of PI3Kβ, defined as MGMT-deficient/PI3Kβ-high, were less responsive to temozolomide and experienced poor prognosis. Consistently, MGMT-deficient/PI3Kβ-high glioblastoma cells were resistant to temozolomide. Perturbation of PI3Kβ, but not other kinases, sensitized MGMT-deficient/PI3Kβ-high glioblastoma cells or tumors to temozolomide. Moreover, PI3Kβ-selective inhibitors and temozolomide synergistically mitigated the growth of glioblastoma stem cells. Our results have demonstrated an essential role of PI3Kβ in chemoresistance, making PI3Kβ-selective blockade an effective chemosensitizer for glioblastoma.
Collapse
Affiliation(s)
- Kevin J. Pridham
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Kasen R. Hutchings
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Patrick Beck
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Min Liu
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Eileen Xu
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Erin Saechin
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016, USA
| | - Vincent Bui
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Chinkal Patel
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Jamie Solis
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Leah Huang
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Allison Tegge
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
| | - Deborah F. Kelly
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802, USA
- Center for Structural Oncology, Pennsylvania State University, University Park, PA 16802, USA
| | - Zhi Sheng
- Fralin Biomedical Research Institute at VTC, Roanoke, VA 24016, USA
- Department of Biomedical Engineering, Pennsylvania State University, University Park, PA 16802, USA
- Faculty of Health Science, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
11
|
Teraiya M, Krokhin O, Chen VC, Perreault H. Cytoplasmic Shotgun Proteomic Points to Key Proteins and Pathways in Temozolomide-Resistant Glioblastoma Multiforme. J Proteome Res 2024; 23:465-482. [PMID: 38147655 DOI: 10.1021/acs.jproteome.3c00669] [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] [Indexed: 12/28/2023]
Abstract
Temozolomide (TMZ) is the first line of chemotherapy to treat primary brain tumors of the type glioblastoma multiforme (GBM). TMZ resistance (TMZR) is one of the main barriers to successful treatment and is a principal factor in relapse, resulting in a poor median survival of 15 months. The present paper focuses on proteomic analyses of cytosolic fractions from TMZ-resistant (TMZR) LN-18 cells. The experimental workflow includes an easy, cost-effective, and reproducible method to isolate subcellular fraction of cytosolic (CYTO) proteins, mitochondria, and plasma membrane proteins for proteomic studies. For this study, enriched cytoplasmic fractions were analyzed in replicates by nanoflow liquid chromatography tandem high-resolution mass spectrometry (nLC-MS/MS), and proteins identified were quantified using a label-free approach (LFQ). Statistical analysis of control (CTRL) and temozolomide-resistant (TMZR) proteomes revealed proteins that appear to be differentially controlled in the cytoplasm. The functions of these proteins are discussed as well as their roles in other cancers and TMZ resistance in GBM. Key proteins are also described through biological processes related to gene ontology (GO), molecular functions, and cellular components. For protein-protein interactions (PPI), network and pathway involvement analyses have been performed, highlighting the roles of key proteins in the TMZ resistance phenotypes. This study provides a detailed insight into methods of subcellular fractionation for proteomic analysis of TMZ-resistant GBM cells and the potential to apply this approach to future large-scale studies. Several key proteins, protein-protein interactions (PPI), and pathways have been identified, underlying the TMZ resistance phenotype and highlighting the proteins' biological functions.
Collapse
Affiliation(s)
- Milan Teraiya
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
| | - Oleg Krokhin
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
- Manitoba Centre for Proteomics and Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba R3E3P4, Canada
| | - Vincent C Chen
- Chemistry Department, Brandon University, Brandon, Manitoba R7A 6A9, Canada
| | - Hélène Perreault
- Chemistry Department, University of Manitoba, Winnipeg, Manitoba R3T3C7, Canada
| |
Collapse
|
12
|
Marangon D, Lecca D. Exosomal non-coding RNAs in glioma progression: insights into tumor microenvironment dynamics and therapeutic implications. Front Cell Dev Biol 2023; 11:1275755. [PMID: 38020906 PMCID: PMC10646304 DOI: 10.3389/fcell.2023.1275755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Gliomas are the most common and deadly types of brain tumors, known for their extensive genetic and epigenetic variability, which poses considerable challenges for pharmacological treatment. Glioma heterogeneity is also related to their intricate and dynamic tumor microenvironment (TME), which comprises a diverse array of cell types, including immune cells, vascular cells, glial cells, and neural precursors, collectively influencing tumor behavior and progression. A pivotal aspect of this intercellular communication relies on the exchange of extracellular vesicles (EVs), which contain and transfer complex molecular cargoes typical of their cells of origin, such as proteins, lipids, carbohydrates, metabolites, and non-coding RNAs (ncRNAs), that encompass microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs). Glioma cells actively release EVs loaded with specific ncRNAs that can target genes and other ncRNAs in recipient cells residing within the TME. Among these recipient cells, prominent players include tumor-associated macrophages and microglia (TAMs), non-neoplastic astrocytes and endothelial cells. The intricate interplay between EVs derived from glioma cells and these recipient cells significantly contributes to the establishment of a tumor-permissive microenvironment, promoting tumor cell proliferation, migration, angiogenesis, and invasion, by targeting various downstream pathways. This review critically examines the current understanding of the intricate interplay between glioma, exosomal ncRNAs, and various components of the glioma TME. By shedding light on the roles of ncRNAs in mediating intercellular communication, this review underscores their significance in orchestrating TME transformation and highlights their potential as novel therapeutic targets for effectively tackling glioma progression.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
13
|
Leske H, Blakstad H, Lund-Iversen M, Skovholt EK, Niehusmann P, Ramm-Pettersen JT, Skogen K, Kongelf G, Sprauten M, Magelssen H, Brandal P. Astrocytoma (CNS WHO grade 4), IDH-mutant with co-occurrence of BRAF p.V600E mutation, and homozygous loss of CDKN2A. Neuropathology 2023; 43:385-390. [PMID: 36754566 DOI: 10.1111/neup.12895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/10/2023]
Abstract
Molecular alterations nowadays play a crucial role in the diagnosis of brain tumors. Some of these alterations are associated with outcome and/or response to treatment, including sequence variants of isocitrate dehydrogenase (IDH) at position p.R132 or p.R172. Such IDH variants have so far been described in histone H3-wildtype primary brain tumors only in adult-type diffuse gliomas and are associated with a better outcome compared to their IDH-wildtype counterpart, the glioblastoma. Moreover, homozygous loss of CDKN2A and/or CDKN2B in IDH-mutant astrocytomas shortens the median overall survival regardless of histological features of malignancy. Such tumors are therefore considered to be aggressive and graded as WHO central nervous system (CNS) grade 4 lesions. The coexistence of an IDH-sequence variation and a BRAF p.V600E alteration has only rarely been described in diffuse astrocytomas. Due to the small number of cases, little is known about such neoplasms in terms of clinical behavior and response to treatment. Herein we describe the first case, to our knowledge, of an astrocytoma (CNS WHO grade 4), IDH-mutant, and BRAF p.V600E-mutant with homozygous deletion of CDKN2A. Pathologists should be aware that such an expression profile does exist even in WHO CNS grade 4 astrocytomas, IDH-mutant, and are encouraged to test for the BRAF p.V600E sequence variant as such an alteration may provide additional treatment options.
Collapse
Affiliation(s)
- Henning Leske
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- University of Oslo (UiO), Oslo, Norway
| | - Hanne Blakstad
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | | | | | - Pitt Niehusmann
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Karoline Skogen
- Department of Radiology, Oslo University Hospital, Oslo, Norway
| | - Geir Kongelf
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Mette Sprauten
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Petter Brandal
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
14
|
Allahyarzadeh Khiabani N, Amin Doustvandi M, Mohammadnejad F, Salmani Hassan Kohal E, Boushehri N, Jafarlou M, Baradaran B. Combination of B7H6-siRNA and temozolomide synergistically reduces stemness and migration properties of glioblastoma cancer cells. Exp Cell Res 2023:113667. [PMID: 37247720 DOI: 10.1016/j.yexcr.2023.113667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/20/2023] [Accepted: 05/25/2023] [Indexed: 05/31/2023]
Abstract
Glioblastoma multiforme (GBM) is among the malignant brain tumors of the central nervous system (CNS). The survival of this disease is about 14 months after diagnosis. To date, temozolomide is known as first-line therapy for glioma. Drug resistance and severe side effects against this drug are important obstacles to the effective treatment of this cancer. Small interfering RNA (siRNA) can adjust the expression of several genes and is used as a new method of gene therapy. Recent studies have shown that siRNAs can increase the sensitivity of cancer cells to chemotherapy drugs. This study aimed to understand the potential role and molecular mechanism of the combination therapy of B7H6-siRNA and temozolomide in glioblastoma cancer. U87 cells were treated with B7H6-siRNA and temozolomide, separately and in combination. Cell viability, stemness, cell migration, and apoptosis were measured. The results of this work presented the synergistic effect of B7H6-siRNA and temozolomide in inhibiting the cancerous features of the U87 cell line. Down-regulating B7H6-siRNA expression inhibited the cell viability of U87 glioblastoma cancer cells and increased their sensitivity to temozolomide. In addition, a noteworthy decrease in cell migration ability and stemness, an increase in apoptosis were observed in the combined groups compared to B7H6-siRNA and temozolomide individually. According to the results, a combination of B7H6-siRNA and temozolomide can be a promising strategy in glioblastoma cancer therapy.
Collapse
Affiliation(s)
- Nadia Allahyarzadeh Khiabani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Genetics, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Mohammad Amin Doustvandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Neda Boushehri
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
15
|
Psidium guajava induces cytotoxicity in human malignant glioblastoma cell line: Role of reactive oxygen species. Toxicol In Vitro 2023; 89:105567. [PMID: 36758825 DOI: 10.1016/j.tiv.2023.105567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/10/2023]
Abstract
One of the deadliest types of CNS primary brain cancers is glioblastoma multiforme (GBM), and the survival rate of patients is about 7.2%. The standard treatment for GBM is surgical interventions followed by temozolomide. We investigated for the first time, the cytotoxic impacts of Psidium guajava (P. guajava) on the U87 GBM cell line. We measured cell toxicity through the MTT test following 24 h, 48 h, and 72 h treatment with different concentrations of fruit and seed hydroalcoholic extracts of P. guajava (25-400 μg/ml). Lipid peroxidation assay, reactive oxygen species (ROS) production, and apoptosis rate were evaluated 24 h after treatment by extracts of P. guajava. Moreover, to determine the Bax/Bcl-2 and NF-κB genes expression, we performed a real-time polymerase chain reaction (RT-PCR). Our finding demonstrated that 50-400 μg/ml of P. guajava extracts dose-dependently decreased the viability of U87 cells. Also, treatment by extracts increased lipid peroxidation, ROS production, and apoptosis in a dose-dependent manner. Moreover, the RT-PCR demonstrated an up-regulation in Bax\Bcl-2 and NF-κB. Thus, P. guajava inhibited the proliferation of U87 GBM cells and increased apoptosis probably through Bax/Bcl-2 and NF-κB regulation.
Collapse
|
16
|
Ali MY, Griguer CE, Flor S, Oliva CR. Mitoferrin-1 Promotes Proliferation and Abrogates Protein Oxidation via the Glutathione Pathway in Glioblastoma. Antioxidants (Basel) 2023; 12:antiox12020349. [PMID: 36829908 PMCID: PMC9952016 DOI: 10.3390/antiox12020349] [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: 01/17/2023] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Median overall survival is very low in patients with glioblastoma (GBM), largely because these tumors become resistant to therapy. Recently, we found that a decrease in the cytosolic labile iron pool underlies the acquisition of radioresistance. Both cytosolic and mitochondrial iron are important for regulating ROS production, which largely facilitates tumor progression and response to therapy. Here, we investigated the role of the mitochondrial iron transporters mitoferrin-1 (MFRN1) and mitoferrin-2 (MFRN2) in GBM progression. Analysis of The Cancer Genome Atlas database revealed upregulation of MFRN1 mRNA and downregulation of MFRN2 mRNA in GBM tumor tissue compared with non-GBM tissue, yet only the tumor expression level of MFRN1 mRNA negatively correlated with overall survival in patients. Overexpression of MFRN1 in glioma cells significantly increased the level of mitochondrial iron, enhanced the proliferation rate and anchorage-independent growth of these cells, and significantly decreased mouse survival in an orthotopic model of glioma. Finally, MFRN1 overexpression stimulated the upregulation of glutathione, which protected glioma cells from 4-hydroxynonenal-induced protein damage. Overall, these results demonstrate a mechanistic link between MFRN1-mediated mitochondrial iron metabolism and GBM progression. Manipulation of MFRN1 may provide a new therapeutic strategy for improving clinical outcomes in patients with GBM.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
| | - Corinne E. Griguer
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Susanne Flor
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
- Free Radical & Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Claudia R. Oliva
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
17
|
Exploring the Past, Present, and Future of Anti-Angiogenic Therapy in Glioblastoma. Cancers (Basel) 2023; 15:cancers15030830. [PMID: 36765787 PMCID: PMC9913517 DOI: 10.3390/cancers15030830] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Glioblastoma, a WHO grade IV astrocytoma, constitutes approximately half of malignant tumors of the central nervous system. Despite technological advancements and aggressive multimodal treatment, prognosis remains dismal. The highly vascularized nature of glioblastoma enables the tumor cells to grow and invade the surrounding tissue, and vascular endothelial growth factor-A (VEGF-A) is a critical mediator of this process. Therefore, over the past decade, angiogenesis, and more specifically, the VEGF signaling pathway, has emerged as a therapeutic target for glioblastoma therapy. This led to the FDA approval of bevacizumab, a monoclonal antibody designed against VEGF-A, for treatment of recurrent glioblastoma. Despite the promising preclinical data and its theoretical effectiveness, bevacizumab has failed to improve patients' overall survival. Furthermore, several other anti-angiogenic agents that target the VEGF signaling pathway have also not demonstrated survival improvement. This suggests the presence of other compensatory angiogenic signaling pathways that surpass the anti-angiogenic effects of these agents and facilitate vascularization despite ongoing VEGF signaling inhibition. Herein, we review the current state of anti-angiogenic agents, discuss potential mechanisms of anti-angiogenic resistance, and suggest potential avenues to increase the efficacy of this therapeutic approach.
Collapse
|
18
|
Song P, Li H, Xu K, Li ZW, Ren X, Fu XJ. A bibliometric and visualization-based analysis of temozolomide research hotspots and frontier evolution. Front Oncol 2022; 12:905868. [DOI: 10.3389/fonc.2022.905868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022] Open
Abstract
The literature related to TMZ research in the Web of Science (WOS) database was analyzed using bibliometrics and visualization by Citespace and VOSviewer.The publication status (number of publications, institutions, and frequency of citations), collaborations, and research focus was analyzed to clarify the current situation of TMZ research. And the recent research on TMZ provides a detailed summary. Based on objective data analysis, this study provides a complete analysis portraying the progression of historical milestones in TMZ development and future research directions from various TMZ research domains.
Collapse
|
19
|
Expression of ABCB1, ABCC1 and 3 and ABCG2 in glioblastoma and their relevance in relation to clinical survival surrogates. J Neurooncol 2022; 160:601-609. [DOI: 10.1007/s11060-022-04179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/21/2022] [Indexed: 11/09/2022]
|
20
|
Chen J, Li Z, Zhao Q, Chen L. Roles of apelin/APJ system in cancer: Biomarker, predictor, and emerging therapeutic target. J Cell Physiol 2022; 237:3734-3751. [DOI: 10.1002/jcp.30845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Jiawei Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| | - Zhiyue Li
- Health Management Center, The Third Xiangya Hospital Central South University Changsha Hunan Province China
| | - Qun Zhao
- Department of Orthopedics Third Xiangya Hospital of Central South University Changsha Hunan China
| | - Linxi Chen
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang Medical School, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy and Pharmacology University of South China Hengyang Hunan China
| |
Collapse
|
21
|
Tumor Cells-derived exosomal CircRNAs: Novel cancer drivers, molecular mechanisms, and clinical opportunities. Biochem Pharmacol 2022; 200:115038. [DOI: 10.1016/j.bcp.2022.115038] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022]
|
22
|
Proto MC, Fiore D, Piscopo C, Laezza C, Bifulco M, Gazzerro P. Modified Adenosines Sensitize Glioblastoma Cells to Temozolomide by Affecting DNA Methyltransferases. Front Pharmacol 2022; 13:815646. [PMID: 35559231 PMCID: PMC9086827 DOI: 10.3389/fphar.2022.815646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal primary malignant brain tumor, and due to its unique features, its management is certainly one of the most challenging ones among all cancers. N6-isopentenyladenosine (IPA) and its analog N6-benzyladenosine (N6-BA) are modified nucleosides endowed with potent antitumor activity on different types of human cancers, including GBM. Corroborating our previous finding, we demonstrated that IPA and N6-BA affect GBM cell line proliferation by modulating the expression of the F-box WD repeat domain-containing-7 (FBXW7), a tumor suppressor with a crucial role in the turnover of many proteins, such as SREBPs and Mcl1, involved in malignant progression and chemoresistance. Luciferase assay revealed that IPA-mediated upregulation of FBXW7 translates in transcriptional inactivation of its oncogenic substrates (Myc, NFkB, or HIF-1α). Moreover, downregulating MGMT expression, IPA strongly enhances the killing effect of temozolomide (TMZ), producing a favorable sensitizing effect starting from a concentration range much lower than TMZ EC50. Through DNA methyltransferase (DNMT) activity assay, analysis of the global DNA methylation, and the histone modification profiles, we demonstrated that the modified adenosines behave similar to 5-AZA-dC, known DNMT inhibitor. Overall, our results provide new perspectives for the first time, suggesting the modified adenosines as epigenetic tools able to improve chemo- and radiotherapy efficacy in glioblastoma and potentially other cancers.
Collapse
Affiliation(s)
| | - Donatella Fiore
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Piscopo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Chiara Laezza
- Institute of Endocrinology and Experimental Oncology, IEOS CNR, Naples, Italy
| | - Maurizio Bifulco
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | | |
Collapse
|
23
|
Abstract
Glioblastoma remains the deadliest form of brain cancer, largely because these tumors become resistant to standard of care treatment with radiation and chemotherapy. Intracellular production of reactive oxygen species (ROS) is necessary for chemo- and radiotherapy-induced cytotoxicity. Here, we assessed whether antioxidant catalase (CAT) affects glioma cell sensitivity to temozolomide and radiation. Using The Cancer Genome Atlas database, we found that CAT mRNA expression is upregulated in glioma tumor tissue compared with non-tumor tissue, and the level of expression negatively correlates with the overall survival of patients with high-grade glioma. In U251 glioma cells, CAT overexpression substantially decreased the basal level of hydrogen peroxide, enhanced anchorage-independent cell growth, and facilitated resistance to the chemotherapeutic drug temozolomide and ionizing radiation. Importantly, pharmacological inhibition of CAT activity reduced the proliferation of glioma cells isolated from patient biopsy samples. Moreover, U251 cells overexpressing CAT formed neurospheres in neurobasal medium, whereas control cells did not, suggesting that the radio- and chemoresistance conferred by CAT may be due in part to the enrichment of glioma stem cell populations. Finally, CAT overexpression significantly decreased survival in an orthotopic mouse model of glioma. These results demonstrate that CAT regulates chemo- and radioresistance in human glioma.
Collapse
|
24
|
Catalase Overexpression Drives an Aggressive Phenotype in Glioblastoma. Antioxidants (Basel) 2021; 10:antiox10121988. [PMID: 34943091 PMCID: PMC8750785 DOI: 10.3390/antiox10121988] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma remains the deadliest form of brain cancer, largely because these tumors become resistant to standard of care treatment with radiation and chemotherapy. Intracellular production of reactive oxygen species (ROS) is necessary for chemo- and radiotherapy-induced cytotoxicity. Here, we assessed whether antioxidant catalase (CAT) affects glioma cell sensitivity to temozolomide and radiation. Using The Cancer Genome Atlas database, we found that CAT mRNA expression is upregulated in glioma tumor tissue compared with non-tumor tissue, and the level of expression negatively correlates with the overall survival of patients with high-grade glioma. In U251 glioma cells, CAT overexpression substantially decreased the basal level of hydrogen peroxide, enhanced anchorage-independent cell growth, and facilitated resistance to the chemotherapeutic drug temozolomide and ionizing radiation. Importantly, pharmacological inhibition of CAT activity reduced the proliferation of glioma cells isolated from patient biopsy samples. Moreover, U251 cells overexpressing CAT formed neurospheres in neurobasal medium, whereas control cells did not, suggesting that the radio- and chemoresistance conferred by CAT may be due in part to the enrichment of glioma stem cell populations. Finally, CAT overexpression significantly decreased survival in an orthotopic mouse model of glioma. These results demonstrate that CAT regulates chemo- and radioresistance in human glioma.
Collapse
|
25
|
Slow Off-Rate Modified Aptamer (SOMAmer) Proteomic Analysis of Patient-Derived Malignant Glioma Identifies Distinct Cellular Proteomes. Int J Mol Sci 2021; 22:ijms22179566. [PMID: 34502484 PMCID: PMC8431317 DOI: 10.3390/ijms22179566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/04/2023] Open
Abstract
Malignant gliomas derive from brain glial cells and represent >75% of primary brain tumors. This includes anaplastic astrocytoma (grade III; AS), the most common and fatal glioblastoma multiforme (grade IV; GBM), and oligodendroglioma (ODG). We have generated patient-derived AS, GBM, and ODG cell models to study disease mechanisms and test patient-centered therapeutic strategies. We have used an aptamer-based high-throughput SOMAscan® 1.3K assay to determine the proteomic profiles of 1307 different analytes. SOMAscan® proteomes of AS and GBM self-organized into closely adjacent proteomes which were clearly distinct from ODG proteomes. GBM self-organized into four proteomic clusters of which SOMAscan® cluster 4 proteome predicted a highly inter-connected proteomic network. Several up- and down-regulated proteins relevant to glioma were successfully validated in GBM cell isolates across different SOMAscan® clusters and in corresponding GBM tissues. Slow off-rate modified aptamer proteomics is an attractive analytical tool for rapid proteomic stratification of different malignant gliomas and identified cluster-specific SOMAscan® signatures and functionalities in patient GBM cells.
Collapse
|
26
|
Delello Di Filippo L, Hofstätter Azambuja J, Paes Dutra JA, Tavares Luiz M, Lobato Duarte J, Nicoleti LR, Olalla Saad ST, Chorilli M. Improving temozolomide biopharmaceutical properties in glioblastoma multiforme (GBM) treatment using GBM-targeting nanocarriers. Eur J Pharm Biopharm 2021; 168:76-89. [PMID: 34461214 DOI: 10.1016/j.ejpb.2021.08.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/24/2021] [Accepted: 08/22/2021] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain cancer. GBM has aggressive development, and the pharmacological treatment remains a challenge due to GBM anatomical characteristics' (the blood-brain barrier and tumor microenvironment) and the increasing resistance to marketed drugs, such as temozolomide (TMZ), the first-line drug for GBM treatment. Due to physical-chemical properties such as short half-life time and the increasing resistance shown by GBM cells, high doses and repeated administrations are necessary, leading to significant adverse events. This review will discuss the main molecular mechanisms of TMZ resistance and the use of functionalized nanocarriers as an efficient and safe strategy for TMZ delivery. GBM-targeting nanocarriers are an important tool for the treatment of GBM, demonstrating to improve the biopharmaceutical properties of TMZ and repurpose its use in anti-GBM therapy. Technical aspects of nanocarriers will be discussed, and biological models highlighting the advantages and effects of functionalization strategies in TMZ anti-GBM activity. Finally, conclusions regarding the main findings will be made in the context of new perspectives for the treatment of GBM using TMZ as a chemotherapy agent, improving the sensibility and biological anti-tumor effect of TMZ through functionalization strategies.
Collapse
Affiliation(s)
| | | | | | - Marcela Tavares Luiz
- School of Pharmaceutical Science of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Jonatas Lobato Duarte
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Luiza Ribeiro Nicoleti
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Sara Teresinha Olalla Saad
- Hematology and Transfusion Medicine Center, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| |
Collapse
|
27
|
Di Giorgio E, Cuocolo A, Mansi L, Sicignano M, Squame F, Gaudieri V, Giordano P, Giugliano FM, Mazzaferro MP, Negro A, Villa A, Spadafora M. Assessment of therapy response to Regorafenib by 18F-DOPA-PET/CT in patients with recurrent high-grade gliomas: a case series. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
28
|
Wang S, Dong Y, Gong A, Kong H, Gao J, Hao X, Liu Y, Wang Z, Fan Y, Liu C, Xu W. Exosomal circRNAs as novel cancer biomarkers: Challenges and opportunities. Int J Biol Sci 2021; 17:562-573. [PMID: 33613113 PMCID: PMC7893596 DOI: 10.7150/ijbs.48782] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/19/2020] [Indexed: 12/14/2022] Open
Abstract
Identifying high specificity and sensitivity biomarkers has always been the focus of research in the field of non-invasive cancer diagnosis. Exosomes are extracellular vesicles with a lipid bilayer membrane that can be released by all types of cells, which contain a variety of proteins, lipids, and a variety of non-coding RNAs. Increasing research has shown that the lipid bilayer can effectively protect the nucleic acid in exosomes. In cancers, tumor cell-derived exosomal circRNAs can act on target cells or organs through the transport of exosomes, and then participate in the regulation of tumor development and metastasis. Since exosomes exist in various body fluids and circRNAs in exosomes exhibit high stability, exosomal circRNAs have the potential as biomarkers for early and minimally invasive cancer diagnosis and prognosis judgment. In this review, we summarized circRNAs and their biological roles in cancers, with the emerging value biomarkers in cancer diagnosis, disease judgment, and prognosis observation. In addition, we briefly compared the advantages of exosomal circRNAs as biomarkers and the current obstacles in the exosome isolation technology, shed light to the future development of this technology.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Yanhan Dong
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Anjing Gong
- Department of Neurosurgery, The affiliated hospital of Qingdao University, Qingdao 266003, China
| | - Huimin Kong
- School Hospital, Shandong University of Science and Technology, Qingdao 266003, China
| | - Jinning Gao
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Xiaodan Hao
- Institute of Translational Medicine, Qingdao University, Qingdao, 266003, China
| | - Yongmei Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Zibo Wang
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Yuqiao Fan
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Chengyu Liu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| | - Wenhua Xu
- Department of Inspection, The medical faculty of Qingdao University, Qingdao 266003, China
| |
Collapse
|
29
|
Yang Y, Okada S, Sakurai M. Adenosine-to-inosine RNA editing in neurological development and disease. RNA Biol 2021; 18:999-1013. [PMID: 33393416 DOI: 10.1080/15476286.2020.1867797] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adenosine-to-inosine (A-to-I) editing is one of the most prevalent post-transcriptional RNA modifications in metazoan. This reaction is catalysed by enzymes called adenosine deaminases acting on RNA (ADARs). RNA editing is involved in the regulation of protein function and gene expression. The numerous A-to-I editing sites have been identified in both coding and non-coding RNA transcripts. These editing sites are also found in various genes expressed in the central nervous system (CNS) and play an important role in neurological development and brain function. Aberrant regulation of RNA editing has been associated with the pathogenesis of neurological and psychiatric disorders, suggesting the physiological significance of RNA editing in the CNS. In this review, we discuss the current knowledge of editing on neurological disease and development.
Collapse
Affiliation(s)
- Yuxi Yang
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba, Japan
| | - Shunpei Okada
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba, Japan
| | - Masayuki Sakurai
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda-shi, Chiba, Japan
| |
Collapse
|
30
|
Molinaro AM, Hervey-Jumper S, Morshed RA, Young J, Han SJ, Chunduru P, Zhang Y, Phillips JJ, Shai A, Lafontaine M, Crane J, Chandra A, Flanigan P, Jahangiri A, Cioffi G, Ostrom Q, Anderson JE, Badve C, Barnholtz-Sloan J, Sloan AE, Erickson BJ, Decker PA, Kosel ML, LaChance D, Eckel-Passow J, Jenkins R, Villanueva-Meyer J, Rice T, Wrensch M, Wiencke JK, Oberheim Bush NA, Taylor J, Butowski N, Prados M, Clarke J, Chang S, Chang E, Aghi M, Theodosopoulos P, McDermott M, Berger MS. Association of Maximal Extent of Resection of Contrast-Enhanced and Non-Contrast-Enhanced Tumor With Survival Within Molecular Subgroups of Patients With Newly Diagnosed Glioblastoma. JAMA Oncol 2020; 6:495-503. [PMID: 32027343 DOI: 10.1001/jamaoncol.2019.6143] [Citation(s) in RCA: 376] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Per the World Health Organization 2016 integrative classification, newly diagnosed glioblastomas are separated into isocitrate dehydrogenase gene 1 or 2 (IDH)-wild-type and IDH-mutant subtypes, with median patient survival of 1.2 and 3.6 years, respectively. Although maximal resection of contrast-enhanced (CE) tumor is associated with longer survival, the prognostic importance of maximal resection within molecular subgroups and the potential importance of resection of non-contrast-enhanced (NCE) disease is poorly understood. Objective To assess the association of resection of CE and NCE tumors in conjunction with molecular and clinical information to develop a new road map for cytoreductive surgery. Design, Setting, and Participants This retrospective, multicenter cohort study included a development cohort from the University of California, San Francisco (761 patients diagnosed from January 1, 1997, through December 31, 2017, with 9.6 years of follow-up) and validation cohorts from the Mayo Clinic (107 patients diagnosed from January 1, 2004, through December 31, 2014, with 5.7 years of follow-up) and the Ohio Brain Tumor Study (99 patients with data collected from January 1, 2008, through December 31, 2011, with a median follow-up of 10.9 months). Image accessors were blinded to patient groupings. Eligible patients underwent surgical resection for newly diagnosed glioblastoma and had available survival, molecular, and clinical data and preoperative and postoperative magnetic resonance images. Data were analyzed from November 15, 2018, to March 15, 2019. Main Outcomes and Measures Overall survival. Results Among the 761 patients included in the development cohort (468 [61.5%] men; median age, 60 [interquartile range, 51.6-67.7] years), younger patients with IDH-wild-type tumors and aggressive resection of CE and NCE tumors had survival similar to that of patients with IDH-mutant tumors (median overall survival [OS], 37.3 [95% CI, 31.6-70.7] months). Younger patients with IDH-wild-type tumors and reduction of CE tumor but residual NCE tumors fared worse (median OS, 16.5 [95% CI, 14.7-18.3] months). Older patients with IDH-wild-type tumors benefited from reduction of CE tumor (median OS, 12.4 [95% CI, 11.4-14.0] months). The results were validated in the 2 external cohorts. The association between aggressive CE and NCE in patients with IDH-wild-type tumors was not attenuated by the methylation status of the promoter region of the DNA repair enzyme O6-methylguanine-DNA methyltransferase. Conclusions and Relevance This study confirms an association between maximal resection of CE tumor and OS in patients with glioblastoma across all subgroups. In addition, maximal resection of NCE tumor was associated with longer OS in younger patients, regardless of IDH status, and among patients with IDH-wild-type glioblastoma regardless of the methylation status of the promoter region of the DNA repair enzyme O6-methylguanine-DNA methyltransferase. These conclusions may help reassess surgical strategies for individual patients with newly diagnosed glioblastoma.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco
| | | | - Ramin A Morshed
- Department of Neurological Surgery, University of California, San Francisco
| | - Jacob Young
- Department of Neurological Surgery, University of California, San Francisco
| | - Seunggu J Han
- Department of Neurological Surgery, Oregon Health Sciences University, Portland
| | - Pranathi Chunduru
- Department of Neurological Surgery, University of California, San Francisco
| | - Yalan Zhang
- Department of Neurological Surgery, University of California, San Francisco
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco.,Department of Pathology, University of California, San Francisco
| | - Anny Shai
- Department of Neurological Surgery, University of California, San Francisco
| | - Marisa Lafontaine
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Jason Crane
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Ankush Chandra
- Department of Neurological Surgery, University of California, San Francisco
| | - Patrick Flanigan
- Department of Neurological Surgery, University of California, San Francisco
| | - Arman Jahangiri
- Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia
| | - Gino Cioffi
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Quinn Ostrom
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - John E Anderson
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Department of Radiology, University Hospitals of Cleveland, Cleveland, Ohio
| | - Chaitra Badve
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Department of Radiology, University Hospitals of Cleveland, Cleveland, Ohio
| | - Jill Barnholtz-Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Research Division, University Hospitals of Cleveland, Cleveland, Ohio
| | - Andrew E Sloan
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio.,Seidman Cancer Center, University Hospitals of Cleveland, Cleveland, Ohio
| | | | | | | | | | | | | | | | - Terri Rice
- Department of Neurological Surgery, University of California, San Francisco
| | - Margaret Wrensch
- Department of Neurological Surgery, University of California, San Francisco
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco
| | - Nancy Ann Oberheim Bush
- Department of Neurological Surgery, University of California, San Francisco.,Department of Neurology, University of California, San Francisco
| | - Jennie Taylor
- Department of Neurological Surgery, University of California, San Francisco.,Department of Neurology, University of California, San Francisco
| | - Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco
| | - Michael Prados
- Department of Neurological Surgery, University of California, San Francisco
| | - Jennifer Clarke
- Department of Neurological Surgery, University of California, San Francisco.,Department of Neurology, University of California, San Francisco
| | - Susan Chang
- Department of Neurological Surgery, University of California, San Francisco
| | - Edward Chang
- Department of Neurological Surgery, University of California, San Francisco
| | - Manish Aghi
- Department of Neurological Surgery, University of California, San Francisco
| | | | - Michael McDermott
- Department of Neurological Surgery, University of California, San Francisco
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco
| |
Collapse
|
31
|
Catacuzzeno L, Sforna L, Esposito V, Limatola C, Franciolini F. Ion Channels in Glioma Malignancy. Rev Physiol Biochem Pharmacol 2020; 181:223-267. [DOI: 10.1007/112_2020_44] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Schönthal AH, Swenson SD, Chen TC, Markland FS. Preclinical studies of a novel snake venom-derived recombinant disintegrin with antitumor activity: A review. Biochem Pharmacol 2020; 181:114149. [PMID: 32663453 DOI: 10.1016/j.bcp.2020.114149] [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] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 12/12/2022]
Abstract
Snake venoms consist of a complex mixture of many bioactive molecules. Among them are disintegrins, which are peptides without enzymatic activity, but with high binding affinity for integrins, transmembrane receptors that function to connect cells with components of the extracellular matrix. Integrin-mediated cell attachment is critical for cell migration and dissemination, as well as for signal transduction pathways involved in cell growth. During tumor development, integrins play key roles by supporting cancer cell proliferation, angiogenesis, and metastasis. The recognition that snake venom disintegrins can block integrin functions has spawned a number of studies to explore their cancer therapeutic potential. While dozens of different disintegrins have been isolated, none of them as yet has undergone clinical evaluation in cancer patients. Among the best-characterized and preclinically most advanced disintegrins is vicrostatin (VCN), a recombinant disintegrin that was rationally designed by fusing 62 N-terminal amino acids derived from the disintegrin contortrostatin with 6 C-terminal amino acids from echistatin, the disintegrins from another snake species. Bacterially produced VCN was shown to target multiple tumor-associated integrins, achieving potent anti-tumor and anti-angiogenic effects in in vitro and in vivo models in the absence of noticeable toxicity. This review will introduce the field of snake venom disintegrins as potential anticancer agents and illustrate the translational development and cancer-therapeutic potential of VCN as an example.
Collapse
Affiliation(s)
- Axel H Schönthal
- Department of Molecular Microbiology and Immunology, Keck School of Medicine (KSOM), University of Southern California (USC), Los Angeles, CA 90089, USA
| | - Stephen D Swenson
- Department of Neurological Surgery, KSOM, USC, Los Angeles, CA 90089, USA; Department of Biochemistry and Molecular Medicine, KSOM, USC, Los Angeles, CA 90089, USA
| | - Thomas C Chen
- Department of Neurological Surgery, KSOM, USC, Los Angeles, CA 90089, USA
| | - Francis S Markland
- Department of Biochemistry and Molecular Medicine, KSOM, USC, Los Angeles, CA 90089, USA.
| |
Collapse
|
33
|
Apelin Controls Angiogenesis-Dependent Glioblastoma Growth. Int J Mol Sci 2020; 21:ijms21114179. [PMID: 32545380 PMCID: PMC7312290 DOI: 10.3390/ijms21114179] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 06/09/2020] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) present with an abundant and aberrant tumor neo-vasculature. While rapid growth of solid tumors depends on the initiation of tumor angiogenesis, GBM also progress by infiltrative growth and vascular co-option. The angiogenic factor apelin (APLN) and its receptor (APLNR) are upregulated in GBM patient samples as compared to normal brain tissue. Here, we studied the role of apelin/APLNR signaling in GBM angiogenesis and growth. By functional analysis of apelin in orthotopic GBM mouse models, we found that apelin/APLNR signaling is required for in vivo tumor angiogenesis. Knockdown of tumor cell-derived APLN massively reduced the tumor vasculature. Additional loss of the apelin signal in endothelial tip cells using the APLN-knockout (KO) mouse led to a further reduction of GBM angiogenesis. Direct infusion of the bioactive peptide apelin-13 rescued the vascular loss-of-function phenotype specifically. In addition, APLN depletion massively reduced angiogenesis-dependent tumor growth. Consequently, survival of GBM-bearing mice was significantly increased when APLN expression was missing in the brain tumor microenvironment. Thus, we suggest that targeting vascular apelin may serve as an alternative strategy for anti-angiogenesis in GBM.
Collapse
|
34
|
Azambuja JH, Schuh RS, Michels LR, Gelsleichter NE, Beckenkamp LR, Lenz GS, de Oliveira FH, Wink MR, Stefani MA, Battastini AMO, Teixeira HF, Braganhol E. CD73 as a target to improve temozolomide chemotherapy effect in glioblastoma preclinical model. Cancer Chemother Pharmacol 2020; 85:1177-1182. [PMID: 32417936 DOI: 10.1007/s00280-020-04077-1] [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] [Received: 02/26/2020] [Accepted: 05/06/2020] [Indexed: 11/28/2022]
Abstract
Glioblastoma is the most devastating primary brain tumor and effective therapies are not available. Treatment is based on surgery followed by radio and chemotherapy with temozolomide (TMZ), but TMZ increases patient survival only by 2 months. CD73, an enzyme responsible for adenosine production, emerges as a target for glioblastoma treatment. Indeed, adenosine causes tumor-promoting actions and CD73 inhibition increases sensitivity to TMZ in vitro. Here, a cationic nanoemulsion to nasal delivery of siRNA CD73 (NE-siRNA CD73) aiming glioblastoma treatment was employed alone or in combination with TMZ. In vitro, two glioblastoma cell lines (C6 and U138MG) with a chemo-resistant profile were used. Treatment alone with NE-siRNA CD73 reduced C6 and U138MG glioma cell viability by 70% and 25%, respectively. On the other hand, when NE-siRNA + TMZ combined treatment was employed, a reduction of 85% and 33% of cell viability was observed. Notably, treatment with NE-siRNA CD73 of glioma-bearing Wistar rats reduced tumor size by 80%, 60% more than the standard chemotherapy with TMZ, but no synergistic or additive effect was observed in vivo. Additionally, NE-siRNA CD73, TMZ or combined therapy decreased adenosine levels in liquor confirming the importance of this nucleoside on in vivo GB growth. Finally, no hemolytic potential was observed. These results suggest that nasal administration of NE-siRNA CD73 exhibits higher antiglioma effect when compared to TMZ. However, no synergistic or additive in vivo was promoted by the therapeutic regimen employed in this study.
Collapse
Affiliation(s)
- J H Azambuja
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil.
| | - R S Schuh
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - L R Michels
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - N E Gelsleichter
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil
| | - L R Beckenkamp
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil
| | - G S Lenz
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil
| | | | - M R Wink
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil
| | - M A Stefani
- Departamento de Morfologia, UFRGS, Porto Alegre, RS, Brazil
| | | | - H F Teixeira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - E Braganhol
- Programa de Pós-Graduação Em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245-Prédio Principal, Porto Alegre, RS, 90050-170, Brazil
| |
Collapse
|
35
|
Ding C, Yi X, Wu X, Bu X, Wang D, Wu Z, Zhang G, Gu J, Kang D. Exosome-mediated transfer of circRNA CircNFIX enhances temozolomide resistance in glioma. Cancer Lett 2020; 479:1-12. [PMID: 32194140 DOI: 10.1016/j.canlet.2020.03.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/14/2022]
Abstract
Development of chemotherapy resistance remains a major obstacle for glioma management. Exosome-mediated transfer of circular RNAs (circRNAs) are being found to have relevance to many human cancers, including glioma. The purpose of this study is to explore the effect and underlying mechanism of exosomal circRNA nuclear factor I X (CircNFIX) on temozolomide (TMZ) chemoresistance in glioma. Our results indicated that exosomal CircNFIX was up-regulated in the serum of TMZ-resistant patients and predicted poor prognosis. Exosomal CircNFIX from TMZ-resistant cells conferred TMZ resistance to recipient sensitive cells through the enhancement of cell migration and invasion and the repression of cell apoptosis under TMZ exposure. CircNFIX directly interacted with miR-132 by binding to miR-132. CircNFIX knockdown enhanced TMZ sensitivity in resistant glioma cells by up-regulating miR-132. Additionally, exosomal CircNFIX promoted tumor growth and its depletion enhanced TMZ sensitivity in glioma cells in vivo. Taken together, our study suggests that exosome-mediated transfer of CircNFIX enhances TMZ resistance in glioma at least partially through sponging miR-132, highlighting a potentially prognostic biomarker and therapeutic target for improving the clinical benefits of TMZ treatment in patients with glioma.
Collapse
Affiliation(s)
- Chenyu Ding
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xuehan Yi
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xiyue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Xingyao Bu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| | - Desheng Wang
- Department of Otolaryngology Head and Neck Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, People's Republic of China
| | - Zanyi Wu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China
| | - Gaoqi Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China
| | - Jianjun Gu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450000, Henan, People's Republic of China.
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350001, Fujian, People's Republic of China.
| |
Collapse
|
36
|
Marchi F, Sahnane N, Cerutti R, Cipriani D, Barizzi J, Stefanini FM, Epistolio S, Cerati M, Balbi S, Mazzucchelli L, Sessa F, Pesce GA, Reinert M, Frattini M. The Impact of Surgery in IDH 1 Wild Type Glioblastoma in Relation With the MGMT Deregulation. Front Oncol 2020; 9:1569. [PMID: 32039032 PMCID: PMC6992596 DOI: 10.3389/fonc.2019.01569] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 12/27/2019] [Indexed: 11/13/2022] Open
Abstract
Object: The treatment of choice in glioblastoma (GBM) is the maximal surgical extent of resection (EOR) followed by adjuvant chemo-radiotherapy. Furthermore, methylguanine-DNA methyltransferase (MGMT) promoter methylation is associated with prolonged overall survival (OS) and progression free survival (PFS). The objective of the present study is correlate the biomolecular aspects in relation with EOR. Materials and methods: We analyzed a series of 116 patients with IDH-1 wild type GBM and different EOR (Gross Total Resection—GTR-, Partial Resection—PR- and Biopsy), treated with adjuvant chemo-radiotherapy. The MGMT status was analyzed in terms of promoter methylation and protein expression. Results: When GTR was possible, OS and PFS were significantly better compared to the other two groups (p = 0.001 and p = 0.035, respectively). MGMT methylation was significantly associated with better OS in the biopsy group (p = 0.022) and better OS and PFS in PR (p = 0.02 and p = 0.012, respectively), but not in the GTR group (p = 0.252 for OS, p = 0.256 for PFS) nor the PFS in the biopsy group (p = 0.259). MGMT protein expression levels do not show any association with OS and PFS, regardless of the type of surgery. Conclusions: Our study confirms the positive association of a safe maximal EOR with better OS and PFS, and indicates a positive prognostic value of MGMT methylation status only in case of the presence of residual tumor tissue. MGMT protein expression seems not to play a clinical role in relation with the type of surgery.
Collapse
Affiliation(s)
- Francesco Marchi
- Service of Neurosurgery, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Lugano, Switzerland
| | - Nora Sahnane
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria-ASST Sette Laghi, Varese, Italy
| | - Roberta Cerutti
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria-ASST Sette Laghi, Varese, Italy
| | - Debora Cipriani
- Service of Neurosurgery, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Lugano, Switzerland
| | | | | | | | - Michele Cerati
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria-ASST Sette Laghi, Varese, Italy
| | - Sergio Balbi
- Division of Neurological Surgery, Department of Biotechnology and Life Sciences, University of Insubria-ASST Sette Laghi, Varese, Italy
| | | | - Fausto Sessa
- Unit of Pathology, Department of Medicine and Surgery, University of Insubria-ASST Sette Laghi, Varese, Italy
| | | | - Michael Reinert
- Service of Neurosurgery, Neurocenter of the Southern Switzerland, Regional Hospital of Lugano, Lugano, Switzerland.,Faculty of Medicine, University of Bern, Bern, Switzerland
| | | |
Collapse
|
37
|
Rabé M, Dumont S, Álvarez-Arenas A, Janati H, Belmonte-Beitia J, Calvo GF, Thibault-Carpentier C, Séry Q, Chauvin C, Joalland N, Briand F, Blandin S, Scotet E, Pecqueur C, Clairambault J, Oliver L, Perez-Garcia V, Nadaradjane A, Cartron PF, Gratas C, Vallette FM. Identification of a transient state during the acquisition of temozolomide resistance in glioblastoma. Cell Death Dis 2020; 11:19. [PMID: 31907355 PMCID: PMC6944699 DOI: 10.1038/s41419-019-2200-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 12/17/2022]
Abstract
Drug resistance limits the therapeutic efficacy in cancers and leads to tumor recurrence through ill-defined mechanisms. Glioblastoma (GBM) are the deadliest brain tumors in adults. GBM, at diagnosis or after treatment, are resistant to temozolomide (TMZ), the standard chemotherapy. To better understand the acquisition of this resistance, we performed a longitudinal study, using a combination of mathematical models, RNA sequencing, single cell analyses, functional and drug assays in a human glioma cell line (U251). After an initial response characterized by cell death induction, cells entered a transient state defined by slow growth, a distinct morphology and a shift of metabolism. Specific genes expression associated to this population revealed chromatin remodeling. Indeed, the histone deacetylase inhibitor trichostatin (TSA), specifically eliminated this population and thus prevented the appearance of fast growing TMZ-resistant cells. In conclusion, we have identified in glioblastoma a population with tolerant-like features, which could constitute a therapeutic target.
Collapse
Affiliation(s)
- Marion Rabé
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Solenne Dumont
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,GenoBiRD, SFR François Bonamy, Université de Nantes, Nantes, France
| | - Arturo Álvarez-Arenas
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Hicham Janati
- Laboratoire Jacques-Louis Lions, Inria, Mamba team and Sorbonne Université, Paris 6, UPMC, Paris, France
| | - Juan Belmonte-Beitia
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Gabriel F Calvo
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | | | - Quentin Séry
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Cynthia Chauvin
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Noémie Joalland
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Floriane Briand
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Stéphanie Blandin
- Plate-Forme MicroPICell, SFR François Bonamy, Université de Nantes, Nantes, France
| | - Emmanuel Scotet
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Claire Pecqueur
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France
| | - Jean Clairambault
- Laboratoire Jacques-Louis Lions, Inria, Mamba team and Sorbonne Université, Paris 6, UPMC, Paris, France
| | - Lisa Oliver
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,CHU Nantes, 44093, Nantes, France
| | - Victor Perez-Garcia
- Department of Mathematics and MôLAB-Mathematical Oncology Laboratory, University of Castilla-la Mancha, Ciudad Real, Spain
| | - Arulraj Nadaradjane
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Pierre-François Cartron
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France.,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France
| | - Catherine Gratas
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,CHU Nantes, 44093, Nantes, France.
| | - François M Vallette
- CRCINA, INSERM, Université d'Angers, Université de Nantes, Nantes, France. .,Laboratoire de Biologie des Cancers et Théranostic, Institut de Cancérologie de l'Ouest-St Herblain, 44805, Saint-Herblain, France.
| |
Collapse
|
38
|
Role of myeloid cells in the immunosuppressive microenvironment in gliomas. Immunobiology 2020; 225:151853. [PMID: 31703822 DOI: 10.1016/j.imbio.2019.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/03/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023]
|
39
|
Liu W, Lin J, Chou Y, Li M, Tsai J. CD44-associated radioresistance of glioblastoma in irradiated brain areas with optimal tumor coverage. Cancer Med 2020; 9:350-360. [PMID: 31746135 PMCID: PMC6943151 DOI: 10.1002/cam4.2714] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 10/31/2019] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) requires radiotherapy (RT) as its definitive management. However, GBM still has a high local recurrence rate even after RT. Cancer stem-like cells (CSCs) might enable GBM to evade irradiation damage and cause therapeutic failure. The optimal RT plan should achieve a planning target volume (PTV) coverage of more than 95% but cannot always meet the requirements. Here, we demonstrate that irradiation with different tumor coverage rates to different brain areas has similar effects on GBM. To retrospectively analyze the relationship between PTV coverage and the survival rate in 26 malignant glioblastoma patients, we established primary cell lines from patient-derived malignant glioblastoma cells with the PTV95 (PTV coverage of more than 95%) program (GBM-MG1 cells) and the Non-PTV95 (poor PTV coverage of less than 95%) program (GBM-MG2 cells). The clinical results of PTV95 and Non-PTV95 showed no difference in the overall survival (OS) rate (P = .390) between the two different levels of PTV coverage. GBM-MG1 (PTV95 program) cells exhibited higher radioresistance than GBM-MG2 (Non-PTV95 program) cells. CD44 promotes radioresistance, CSC properties, angiogenesis and cell proliferation in GBM-MG1 (PTV95 program) cells. GBM patients receiving RT with the PTV95 program exhibited higher radioresistance, CSC properties, angiogenesis and cell proliferation than GBM patients receiving RT with the Non-PTV95 program. Moreover, CD44 plays a crucial role in these properties of GBM patients with the PTV95 program.
Collapse
Affiliation(s)
- Wei‐Hsiu Liu
- Department of Neurological SurgeryTri‐Service General Hospital and National Defense Medical CenterTaipeiTaiwanROC
- Department of SurgerySchool of MedicineNational Defense Medical CenterTaipeiTaiwanROC
| | - Jang‐Chun Lin
- Graduate Institute of Clinical MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwanROC
- Department of Radiation OncologyShuang Ho HospitalTaipei Medical UniversityTaipei CityTaiwanROC
- Department of RadiologySchool of MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwanROC
| | - Yu‐Ching Chou
- School of Public HealthNational Defense Medical CenterTaipeiTaiwanROC
| | - Ming‐Hsien Li
- Department of Radiation OncologyShuang Ho HospitalTaipei Medical UniversityTaipei CityTaiwanROC
| | - Jo‐Ting Tsai
- Department of Radiation OncologyShuang Ho HospitalTaipei Medical UniversityTaipei CityTaiwanROC
- Department of RadiologySchool of MedicineCollege of MedicineTaipei Medical UniversityTaipeiTaiwanROC
| |
Collapse
|
40
|
Azambuja JH, Ludwig N, Yerneni S, Rao A, Braganhol E, Whiteside TL. Molecular profiles and immunomodulatory activities of glioblastoma-derived exosomes. Neurooncol Adv 2020; 2:vdaa056. [PMID: 32642708 PMCID: PMC7262743 DOI: 10.1093/noajnl/vdaa056] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glioblastoma is one of the most immunosuppressive human tumors. Emerging data suggest that glioblastoma-derived exosomes (GBex) reprogram the tumor microenvironment into a tumor-promoting milieu by mechanisms that not yet understood. METHODS Exosomes were isolated from supernatants of glioblastoma cell lines by size exclusion chromatography. The GBex endosomal origin, size, protein cargos, and ex vivo effects on immune cell functions were determined. GBex were injected intravenously into mice to evaluate their ability to in vivo modulate normal immune cell subsets. RESULTS GBex carried immunosuppressive proteins, including FasL, TRAIL, CTLA-4, CD39, and CD73, but contained few immunostimulatory proteins. GBex co-incubated with primary human immune cells induced simultaneous activation of multiple molecular pathways. In CD8+ T cells, GBex suppressed TNF-α and INF-γ release and mediated apoptosis. GBex suppressed natural killer (NK) and CD4+ T-cell activation. GBex activated the NF-κB pathway in macrophages and promoted their differentiation into M2 cells. Inhibition of the NF-κB pathway in macrophages reversed the GBex-mediated effects. GBex-driven reprogramming of macrophages involved the release of soluble factors that promoted tumor proliferation in vitro. In mice injected with GBex, the frequency of splenic CD8+ T cells, NK cells, and M1-like macrophages was reduced, while that of naïve and M2-like macrophages increased (P < .05). CONCLUSIONS GBex reprogrammed functions of all types of immune cells in vitro and altered their frequency in vivo. By creating and sustaining a highly immunosuppressive environment, GBex play a key role in promoting tumor progression.
Collapse
Affiliation(s)
- Juliana Hofstatter Azambuja
- Postgraduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Nils Ludwig
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Aparna Rao
- Department of Neurological Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Elizandra Braganhol
- Postgraduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
41
|
Gangemi V, Mignogna C, Guzzi G, Lavano A, Bongarzone S, Cascini GL, Sabatini U. Impact of [ 64Cu][Cu(ATSM)] PET/CT in the evaluation of hypoxia in a patient with Glioblastoma: a case report. BMC Cancer 2019; 19:1197. [PMID: 31810452 PMCID: PMC6898918 DOI: 10.1186/s12885-019-6368-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 11/18/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Glioblastoma multiform (GBM), a malignant brain tumour, has a very often poor prognosis. The therapeutic approach is represented by surgery followed by radiotherapy and chemotherapy. Hypoxia is a factor that causes a reduction of both radiotherapy and chemotherapy effectiveness in GBM and other cancers. Through the use of [64Cu][Cu(ATSM)], a hypoxia-targeting positron emission tomography (PET) radiotracer, is possible to identify the presence of hypoxic areas within a lesion and therefore modulate the therapeutic approach according to the findings. CASE PRESENTATION In this case report, we observed an increase of radiotracer uptake from early acquisition to late acquisition in hypoxia sites and high correlation between [64Cu][Cu(ATSM) PET/CT results and expression of the hypoxia marker HIF-1α. CONCLUSIONS [64Cu][Cu(ATSM) PET/CT represents a valid opportunity to reveal in vivo hypoxic areas in GBM lesion which can guide clinicians on selecting GMB patient's therapeutic scheme.
Collapse
Affiliation(s)
- Vincenzo Gangemi
- Department of Diagnostic Imaging, Nuclear Medicine Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy.
| | - Chiara Mignogna
- Health Science - Interdipartimental Service Center, University "Magna Graecia" of Catanzaro Medical School, Catanzaro, Italy
| | - Giusy Guzzi
- Department of Neurosurgery, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Angelo Lavano
- Department of Neurosurgery, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Salvatore Bongarzone
- School of Biomedical Engineering & Imaging Sciences, King's College London, King's Health Partners, St Thomas' Hospital, London, SE1 7EH, UK
| | - Giuseppe Lucio Cascini
- Department of Diagnostic Imaging, Nuclear Medicine Unit, Magna Graecia University of Catanzaro, Catanzaro, Italy
| | - Umberto Sabatini
- Neuroradiology Unit, University "Magna Graecia", Catanzaro, Italy
| |
Collapse
|
42
|
LINC00174 down-regulation decreases chemoresistance to temozolomide in human glioma cells by regulating miR-138-5p/SOX9 axis. Hum Cell 2019; 33:159-174. [PMID: 31713817 DOI: 10.1007/s13577-019-00281-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023]
Abstract
Temozolomide (TMZ) is one of the most common drugs selected for glioma chemotherapy, but the therapeutic effect of glioma treatment is usually limited due to its resistance. Long non-coding RNA (lncRNA) is gradually found to be a vital regulator in numerous physiological and pathological processes. Lately, it was revealed that LINC00174 could promote CRC cell growth. However, the function and potential regulatory manner of LINC00174 in glioma remain unclear. Our results demonstrated that the expression level of LINC00174 was higher in glioma tissues, and LINC00174 down-regulation could remarkably prevent cell proliferation and promote cell apoptosis in both glioma cells and TMZ-resistant glioma cells. Mechanistic studies revealed that LINC00174 can sponge microRNA-138-5p (miR-138-5p) and down-regulate its expression, thereby up-regulating the protein level of miR-138-5p's target, sex-determining region Y (SRY)-box9 protein (SOX9). Additionally, in vivo experiments revealed that LINC00174 shRNA can serve as a tumor suppressor through down-regulating SOX9 in glioma. In this study, a novel established regulatory way of LINC00174/miR-138-5p/SOX9 axis was systematically studied, which may provide a new manner for glioma therapy.
Collapse
|
43
|
Kwon YM, Je JY, Cha SH, Oh Y, Cho WH. Synergistic combination of chemo‑phototherapy based on temozolomide/ICG‑loaded iron oxide nanoparticles for brain cancer treatment. Oncol Rep 2019; 42:1709-1724. [PMID: 31436296 PMCID: PMC6775808 DOI: 10.3892/or.2019.7289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
Chemo‑photothermal therapy for cancer treatment has received increasing attention due to its selective therapeutic effects. In the present study, the anticancer effects of drug‑loaded Fe3O4 magnetic nanoparticles (MNPs) by chemo‑photothermal therapy on U‑87 MG human glioblastoma cells was investigated. Anticancer drug‑loaded Fe3O4 MNPs were prepared by loading temozolomide (TMZ) and indocyanine green (ICG), and were characterized by X‑ray diffraction, UV‑vis spectroscopy, thermal gravimetric analysis, transmission electron microscope, as well as drug‑loading capacity. Following treatment with near‑infrared (NIR) light irradiation, the administration of Fe3O4‑TMZ‑ICG MNPs resulted in the apoptosis of U‑87 MG glioblastoma cells through the generation of reactive oxygen species. Western blot analysis and reverse transcription‑quantitative polymerase chain reaction revealed that Fe3O4‑TMZ‑ICG MNPs with NIR laser irradiation lead to significantly enhanced anticancer effects on U‑87 MG glioblastoma cells through the modulation of intrinsic and extrinsic apoptosis genes, including Bcl‑2‑associated X protein, Bcl‑2, cytochrome c, caspase‑3, Fas associated via death domain and caspase‑8. These results suggest that Fe3O4‑TMZ‑ICG MNPs may be potential candidates when administered as chemo‑phototherapy for the treatment of brain cancer.
Collapse
Affiliation(s)
- Young Min Kwon
- Department of Neurosurgery, Dong-A University College of Medicine and Dong-A Medical Center, Busan 49201, Republic of Korea
| | - Jae-Young Je
- Department of Marine-Bio Convergence Science, Pukyong National University, Busan 48547, Republic of Korea
| | - Seung Heon Cha
- Department of Neurosurgery and Medical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan 49241, Republic of Korea
| | - Yunok Oh
- Department of Marine-Bio Convergence Science, Pukyong National University, Busan 48547, Republic of Korea
| | - Won Ho Cho
- Department of Neurosurgery and Medical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan 49241, Republic of Korea
| |
Collapse
|
44
|
Fang N, Wu Z, Wang X, Kang D, Li L, Chen Y, Zheng X, Cai S, Liu X, Chen Z, Tu H, Lin Y, Chen J. Automatic and label-free identification of blood vessels in gliomas using the combination of multiphoton microscopy and image analysis. JOURNAL OF BIOPHOTONICS 2019; 12:e201900006. [PMID: 30868750 DOI: 10.1002/jbio.201900006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/19/2019] [Accepted: 03/12/2019] [Indexed: 06/09/2023]
Abstract
Currently, the targeted treatment of tumor based on the tumor microenvironment is newly developed. Blood vessels are the key parts in the tumor microenvironment, which is taken as a new visible target for tumor therapy. Multiphoton microscopy (MPM), based on the second harmonic generation and two-photon excited fluorescence, is available to make the label-free analysis on the blood vessels in human gliomas. MPM can reveal the vascular morphological characteristics in gliomas, including vascular malformation, intense vascular proliferation, perivascular collagen deposition, perivascular lymphocytes aggregation and microvascular proliferation. In addition, the image analysis algorithms were developed to automatically calculate the perivascular collagen content, vascular cavity area, lumen area, wall area and vessel number. Thus, the vascular morphology, the perivascular collagen deposition and intense vascular proliferation degree can be further quantitatively characterized. Compared with the pathological analysis, the combination of MPM and image analysis has potential advantages in making a quantitative and qualitative analyzing on vascular morphology in glioma microenvironment. As micro-endoscope and two-photon fiberscope are technologically improved, this combined method will be a useful imaging way to make the real-time research on the targeting tumor microenvironment in gliomas.
Collapse
Affiliation(s)
- Na Fang
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, People's Republic of China
| | - Zanyi Wu
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Xingfu Wang
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Dezhi Kang
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Lianhuang Li
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, People's Republic of China
| | - Yupeng Chen
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Xianying Zheng
- Department of Radiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Shanshan Cai
- Department of Pathology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, People's Republic of China
| | - Xueyong Liu
- Department of Pathology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Zhida Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, People's Republic of China
| | - Haohua Tu
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Yuanxiang Lin
- Department of Neurosurgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Jianxin Chen
- Key Laboratory of OptoElectronic Science and Technology for Medicine of Ministry of Education, Fujian Provincial Key Laboratory of Photonics Technology, Fujian Normal University, Fuzhou, People's Republic of China
| |
Collapse
|
45
|
Dufort S, Appelboom G, Verry C, Barbier EL, Lux F, Bräuer-Krisch E, Sancey L, Chang SD, Zhang M, Roux S, Tillement O, Le Duc G. Ultrasmall theranostic gadolinium-based nanoparticles improve high-grade rat glioma survival. J Clin Neurosci 2019; 67:215-219. [DOI: 10.1016/j.jocn.2019.05.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/28/2019] [Accepted: 05/27/2019] [Indexed: 11/29/2022]
|
46
|
Azambuja JH, Schuh RS, Michels LR, Gelsleichter NE, Beckenkamp LR, Iser IC, Lenz GS, de Oliveira FH, Venturin G, Greggio S, daCosta JC, Wink MR, Sevigny J, Stefani MA, Battastini AMO, Teixeira HF, Braganhol E. Nasal Administration of Cationic Nanoemulsions as CD73-siRNA Delivery System for Glioblastoma Treatment: a New Therapeutical Approach. Mol Neurobiol 2019; 57:635-649. [DOI: 10.1007/s12035-019-01730-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
|
47
|
Molinaro AM, Taylor JW, Wiencke JK, Wrensch MR. Genetic and molecular epidemiology of adult diffuse glioma. Nat Rev Neurol 2019; 15:405-417. [PMID: 31227792 PMCID: PMC7286557 DOI: 10.1038/s41582-019-0220-2] [Citation(s) in RCA: 478] [Impact Index Per Article: 79.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
The WHO 2007 glioma classification system (based primarily on tumour histology) resulted in considerable interobserver variability and substantial variation in patient survival within grades. Furthermore, few risk factors for glioma were known. Discoveries over the past decade have deepened our understanding of the molecular alterations underlying glioma and have led to the identification of numerous genetic risk factors. The advances in molecular characterization of glioma have reframed our understanding of its biology and led to the development of a new classification system for glioma. The WHO 2016 classification system comprises five glioma subtypes, categorized by both tumour morphology and molecular genetic information, which led to reduced misclassification and improved consistency of outcomes within glioma subtypes. To date, 25 risk loci for glioma have been identified and several rare inherited mutations that might cause glioma in some families have been discovered. This Review focuses on the two dominant trends in glioma science: the characterization of diagnostic and prognostic tumour markers and the identification of genetic and other risk factors. An overview of the many challenges still facing glioma researchers is also included.
Collapse
Affiliation(s)
- Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| | - Jennie W Taylor
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Margaret R Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Institute of Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
48
|
Lin JC, Tsai JT, Chao TY, Ma HI, Liu WH. Musashi-1 Enhances Glioblastoma Migration by Promoting ICAM1 Translation. Neoplasia 2019; 21:459-468. [PMID: 30959276 PMCID: PMC6453839 DOI: 10.1016/j.neo.2019.02.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 02/28/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma multiforme (GBM) is a lethal brain tumor with a mean survival time of 1 year. One major reason for therapeutic failure is that GBM cells have an extraordinary capacity to invade normal brain tissue beyond the surgical margin, accounting for the lack of treatment efficacy. GBM cells that can infiltrate into the healthy brain possess tumor properties of stemness and invasion, and previous studies demonstrate that Musashi-1 (MSI1), a neural stem cell marker, plays an important role in the maintenance of stem cell status, cellular differentiation, and tumorigenesis in cancers. By analyzing neuronal progenitor cell markers and stemness genes, we predicted that MSI1 might be an important factor in GBM pathogenesis. Because inflammation aids in the proliferation and survival of malignant cells, the inflammatory microenvironment also promotes GBM invasion, and intercellular adhesion molecule-1 (ICAM1), a member of the immunoglobulin superfamily, is involved in inflammation. Our results indicate that the above phenomena are likely due to MSI1 upregulation, which occurred simultaneously with higher expression of ICAM1 in GBM cells. Indeed, MSI1 knockdown effectively suppressed ICAM1 expression and blocked GBM cell motility and invasion, whereas overexpressing ICAM1 reversed these effects. According to RNA immunoprecipitation assays, MSI1-mediated mRNA interactions promote ICAM1 translation. Finally, immunohistochemical analysis showed MSI1 and ICAM-1 to be coexpressed at high levels in GBM tissues. Thus, the MSI1/ICAM1 pathway plays an important role in oncogenic resistance, including increased tumor invasion, and MSI1/ICAM1 may be a target for GBM treatment.
Collapse
Affiliation(s)
- Jang-Chun Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Jo-Ting Tsai
- Department of Radiation Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC; Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC
| | - Tsu-Yi Chao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC; Division of Hematology/Oncology, Shuang-Ho Hospital, Taipei Medical University, Taipei, Taiwan, ROC
| | - Hsin-I Ma
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, No.325, Sec. 2, Cheng-Kung Road, Taipei 11490, Taiwan; Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Hsiu Liu
- Department of Neurological Surgery, Tri-Service General Hospital and National Defense Medical Center, No.325, Sec. 2, Cheng-Kung Road, Taipei 11490, Taiwan; Department of Surgery, School of Medicine, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
49
|
Radiation Increases Functional KCa3.1 Expression and Invasiveness in Glioblastoma. Cancers (Basel) 2019; 11:cancers11030279. [PMID: 30813636 PMCID: PMC6468446 DOI: 10.3390/cancers11030279] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 01/25/2019] [Accepted: 02/20/2019] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma (GBM) is a deadly brain tumor, with fast recurrence even after surgical intervention, radio- and chemotherapies. One of the reasons for relapse is the early invasion of surrounding brain parenchyma by GBM, rendering tumor eradication difficult. Recent studies demonstrate that, in addition to eliminate possible residual tumoral cells after surgery, radiation stimulates the infiltrative behavior of GBM cells. The intermediate conductance of Ca2+-activated potassium channels (KCa3.1) play an important role in regulating the migration of GBM. Here, we show that high dose radiation of patient-derived GBM cells increases their invasion, and induces the transcription of key genes related to these functions, including the IL-4/IL-4R pair. In addition, we demonstrate that radiation increases the expression of KCa3.1 channels, and that their pharmacological inhibition counteracts the pro-invasive phenotype induced by radiation in tumor cells. Our data describe a possible approach to treat tumor resistance that follows radiation therapy in GBM patients.
Collapse
|
50
|
The STAT3/Slug Axis Enhances Radiation-Induced Tumor Invasion and Cancer Stem-like Properties in Radioresistant Glioblastoma. Cancers (Basel) 2018; 10:cancers10120512. [PMID: 30551687 PMCID: PMC6315497 DOI: 10.3390/cancers10120512] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 12/07/2018] [Accepted: 12/11/2018] [Indexed: 01/06/2023] Open
Abstract
Glioblastoma multiforme (GBM) requires radiotherapy (RT) as a part of definitive management strategy. RT is highly effective, destroying cancer cells that may exist around the surgical tumor bed. However, GBM still has a poor prognosis and a high local recurrence rate after RT. Accumulating research indicates that GBM contains cancer stem-like cells (CSCs), which are radioresistant and result in therapeutic failure. Additionally, GBM cells can aggressively invade normal brain tissue, inducing therapeutic failure. Using clinical observations, we evaluated the effect of radiation on tumor control. We also explored the biomolecular pathways that connect radioresistance and CSC- and epithelial-mesenchymal transition (EMT)-associated phenotypes in patient-derived GBM cells. Transwell and microarray assay demonstrated that radioresistant GBM cells (GBM-R2I2) exhibit increased invasion and self-renewal abilities compared with parental GBM cells. Finally, to identify potential mechanisms underlying these observations, we used a PCR array to search for molecular markers of cell motility. Signal transducer and activator of transcription 3 (STAT3) directly bound to the Slug promoter in a chromatin immunoprecipitation assay. Reduced STAT3 decreased Slug expression and suppressed cell invasion in GBM-R2I2 cells while increasing Slug reversed these effects. In addition, STAT3 knockdown significantly inhibited CSC properties, synergistically increased the radiotherapeutic effect, and effectively increased the survival rate in vivo. We deciphered a new pathway of GBM radioresistance, invasion, and recurrence via the STAT3/Slug axis that could be a new target of GBM therapy.
Collapse
|