1
|
Nicol C, Pinkham MB, Foote M, McBean A, Conlon E, Ownsworth T. Patient reported outcomes and short-term adjustment trajectories following gamma knife radiosurgery for benign brain tumor. Disabil Rehabil 2024:1-10. [PMID: 39263895 DOI: 10.1080/09638288.2024.2401631] [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: 01/30/2024] [Revised: 08/25/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE Patient reported outcomes (PROs) in the context of Gamma Knife Stereotactic Radiosurgery (GKSRS) for benign brain tumor have been under-researched. This study examined changes in PROs and adjustment trajectories post-GKSRS. METHOD 50 adults (54% female) aged on average 53.18 (SD = 14.76) years with benign brain tumor were assessed 1 week before GKSRS, 1-2 weeks post-GKSRS, and at 3-month follow-up. Telephone-based questionnaires of anxiety and depressive symptoms, cognitive function, symptom burden, and health-related quality of life (HRQoL) were completed. RESULTS Significant improvements in HRQoL, perceived cognitive ability, anxiety, and total brain tumor symptoms were evident between pre-GKSRS and 3-month follow-up. Conversely, there was a significant short-term increase in depressive symptoms at post-GKSRS; however, levels did not differ from pre-GKSRS at follow-up. No significant changes were evident on PROs of headaches or fatigue. About half of the participants (46-51%) experienced reliable improvement in global HRQoL, and one-third (31-34%) reported improved anxiety symptoms. Increased depressive symptoms was seen in 34% of participants post-GKSRS and 18% at follow-up. CONCLUSIONS At 3 months post-GKSRS, improvements in HRQoL, anxiety, perceived cognitive ability, and total brain tumor symptoms were evident. Routine monitoring and support for pre-GKSRS anxiety and depressive symptoms post-GKSRS is recommended.
Collapse
Affiliation(s)
- Chelsea Nicol
- School of Applied Psychology, Griffith University, Mount Gravatt, Australia
- The Hopkins Centre, Menzies Health Institute of Queensland, Griffith University, Mount Gravatt, Australia
| | - Mark B Pinkham
- School of Medicine, University of Queensland, Brisbane, Australia
- Gamma Knife Centre of Queensland, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Matthew Foote
- School of Medicine, University of Queensland, Brisbane, Australia
- Gamma Knife Centre of Queensland, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Angela McBean
- Gamma Knife Centre of Queensland, Princess Alexandra Hospital, Woolloongabba, Australia
| | - Elizabeth Conlon
- School of Applied Psychology, Griffith University, Mount Gravatt, Australia
| | - Tamara Ownsworth
- School of Applied Psychology, Griffith University, Mount Gravatt, Australia
- The Hopkins Centre, Menzies Health Institute of Queensland, Griffith University, Mount Gravatt, Australia
| |
Collapse
|
2
|
Khalighi S, Reddy K, Midya A, Pandav KB, Madabhushi A, Abedalthagafi M. Artificial intelligence in neuro-oncology: advances and challenges in brain tumor diagnosis, prognosis, and precision treatment. NPJ Precis Oncol 2024; 8:80. [PMID: 38553633 PMCID: PMC10980741 DOI: 10.1038/s41698-024-00575-0] [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: 10/19/2023] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
This review delves into the most recent advancements in applying artificial intelligence (AI) within neuro-oncology, specifically emphasizing work on gliomas, a class of brain tumors that represent a significant global health issue. AI has brought transformative innovations to brain tumor management, utilizing imaging, histopathological, and genomic tools for efficient detection, categorization, outcome prediction, and treatment planning. Assessing its influence across all facets of malignant brain tumor management- diagnosis, prognosis, and therapy- AI models outperform human evaluations in terms of accuracy and specificity. Their ability to discern molecular aspects from imaging may reduce reliance on invasive diagnostics and may accelerate the time to molecular diagnoses. The review covers AI techniques, from classical machine learning to deep learning, highlighting current applications and challenges. Promising directions for future research include multimodal data integration, generative AI, large medical language models, precise tumor delineation and characterization, and addressing racial and gender disparities. Adaptive personalized treatment strategies are also emphasized for optimizing clinical outcomes. Ethical, legal, and social implications are discussed, advocating for transparency and fairness in AI integration for neuro-oncology and providing a holistic understanding of its transformative impact on patient care.
Collapse
Affiliation(s)
- Sirvan Khalighi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kartik Reddy
- Department of Radiology, Emory University, Atlanta, GA, USA
| | - Abhishek Midya
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Krunal Balvantbhai Pandav
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA.
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA.
- The Cell and Molecular Biology Program, Winship Cancer Institute, Atlanta, GA, USA.
| |
Collapse
|
3
|
Tran S, Lapidus A, Neal A, Peters KB, Gately L, Ameratunga M. A systematic review of the impact of brain tumours on risk of motor vehicle crashes. J Neurooncol 2024; 166:395-405. [PMID: 38321326 PMCID: PMC10876497 DOI: 10.1007/s11060-024-04586-6] [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: 12/26/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
PURPOSE Brain tumours are associated with neurocognitive impairments that are important for safe driving. Driving is vital to maintaining patient autonomy, despite this there is limited research on driving capacity amongst patients with brain tumours. The purpose of this review is to examine MVC risk in patients with brain tumours to inform development of clearer driving guidelines. METHODS A systematic review was performed using Medline and EMBASE. Observational studies were included. The outcome of interest was MVC or measured risk of MVC in patients with benign or malignant brain tumours. Descriptive analysis and synthesis without meta-analysis were used to summarise findings. A narrative review of driving guidelines from Australia, United Kingdom and Canada was completed. RESULTS Three studies were included in this review. One cohort study, one cross-sectional study and one case-control study were included (19,135 participants) across United States and Finland. One study evaluated the incidence of MVC in brain tumour patients, revealing no difference in MVC rates. Two studies measured MVC risk using driving simulation and cognitive testing. Patients found at higher risk of MVC had greater degrees of memory and visual attention impairments. However, predictive patient and tumour characteristics of MVC risk were heterogeneous across studies. Overall, driving guidelines had clear recommendations on selected conditions like seizures but were vague surrounding neurocognitive deficits. CONCLUSION Limited data exists regarding driving behaviour and MVC incidence in brain tumour patients. Existing guidelines inadequately address neurocognitive complexities in this group. Future studies evaluating real-world data is required to inform development of more applicable driving guidelines. SYSTEMATIC REVIEW REGISTRATION NUMBER PROSPERO 2023 CRD42023434608.
Collapse
Affiliation(s)
- Sophie Tran
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
| | - Adam Lapidus
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
| | - Andrew Neal
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | | | - Lucy Gately
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
- Walter and Eliza Hall Institute for Medical Research, The University of Melbourne, Melbourne, VIC, Australia
| | - Malaka Ameratunga
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia.
- Central Clinical School, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
4
|
Yang Z, Sun L, Chen H, Sun C, Xia L. New progress in the treatment of diffuse midline glioma with H3K27M alteration. Heliyon 2024; 10:e24877. [PMID: 38312649 PMCID: PMC10835306 DOI: 10.1016/j.heliyon.2024.e24877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 02/06/2024] Open
Abstract
Diffuse midline glioma with H3K27 M alteration is a primary malignant tumor located along the linear structure of the brain, predominantly manifesting in children and adolescents. The mortality rate is exceptionally high, with a mere 1 % 5-year survival rate for newly diagnosed patients. Beyond conventional surgery, radiotherapy, and chemotherapy, novel approaches are imperative to enhance patient prognosis. This article comprehensively reviews current innovative treatment modalities and provides updates on the latest research advancements in preclinical studies and clinical trials focusing on H3K27M-altered diffuse midline glioma. The goal is to contribute positively to clinical treatment strategies.
Collapse
Affiliation(s)
- Zhi Yang
- Department of Neurosurgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, WenZhou, 325035, Zhejiang province, China
| | - Liang Sun
- Department of Neurosurgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, WenZhou, 325035, Zhejiang province, China
| | - Haibin Chen
- Department of Neurosurgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, WenZhou, 325035, Zhejiang province, China
| | - Caixing Sun
- Department of Neurosurgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, WenZhou, 325035, Zhejiang province, China
| | - Liang Xia
- Department of Neurosurgery, Cancer Hospital of University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, WenZhou, 325035, Zhejiang province, China
| |
Collapse
|
5
|
Li J, Zhang P, Qu L, Sun T, Duan Y, Wu M, Weng J, Li Z, Gong X, Liu X, Wang Y, Jia W, Su X, Yue Q, Li J, Zhang Z, Barkhof F, Huang RY, Chang K, Sair H, Ye C, Zhang L, Zhuo Z, Liu Y. Deep Learning for Noninvasive Assessment of H3 K27M Mutation Status in Diffuse Midline Gliomas Using MR Imaging. J Magn Reson Imaging 2023; 58:850-861. [PMID: 36692205 DOI: 10.1002/jmri.28606] [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: 10/21/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Determination of H3 K27M mutation in diffuse midline glioma (DMG) is key for prognostic assessment and stratifying patient subgroups for clinical trials. MRI can noninvasively depict morphological and metabolic characteristics of H3 K27M mutant DMG. PURPOSE This study aimed to develop a deep learning (DL) approach to noninvasively predict H3 K27M mutation in DMG using T2-weighted images. STUDY TYPE Retrospective and prospective. POPULATION For diffuse midline brain gliomas, 341 patients from Center-1 (27 ± 19 years, 184 males), 42 patients from Center-2 (33 ± 19 years, 27 males) and 35 patients (37 ± 18 years, 24 males). For diffuse spinal cord gliomas, 133 patients from Center-1 (30 ± 15 years, 80 males). FIELD STRENGTH/SEQUENCE 5T and 3T, T2-weighted turbo spin echo imaging. ASSESSMENT Conventional radiological features were independently reviewed by two neuroradiologists. H3 K27M status was determined by histopathological examination. The Dice coefficient was used to evaluate segmentation performance. Classification performance was evaluated using accuracy, sensitivity, specificity, and area under the curve. STATISTICAL TESTS Pearson's Chi-squared test, Fisher's exact test, two-sample Student's t-test and Mann-Whitney U test. A two-sided P value <0.05 was considered statistically significant. RESULTS In the testing cohort, Dice coefficients of tumor segmentation using DL were 0.87 for diffuse midline brain and 0.81 for spinal cord gliomas. In the internal prospective testing dataset, the predictive accuracies, sensitivities, and specificities of H3 K27M mutation status were 92.1%, 98.2%, 82.9% in diffuse midline brain gliomas and 85.4%, 88.9%, 82.6% in spinal cord gliomas. Furthermore, this study showed that the performance generalizes to external institutions, with predictive accuracies of 85.7%-90.5%, sensitivities of 90.9%-96.0%, and specificities of 82.4%-83.3%. DATA CONCLUSION In this study, an automatic DL framework was developed and validated for accurately predicting H3 K27M mutation using T2-weighted images, which could contribute to the noninvasive determination of H3 K27M status for clinical decision-making. EVIDENCE LEVEL 2 Technical Efficacy: Stage 2.
Collapse
Affiliation(s)
- Junjie Li
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Peng Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Liying Qu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Ting Sun
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yunyun Duan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Minghao Wu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jinyuan Weng
- Department of Medical Imaging Product, Neusoft, Group Ltd., Shenyang, People's Republic of China
| | - Zhaohui Li
- BioMind Inc., Beijing, People's Republic of China
| | - Xiaodong Gong
- Department of Medical Imaging Product, Neusoft, Group Ltd., Shenyang, People's Republic of China
| | - Xing Liu
- Department of Pathology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yongzhi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wenqing Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Xiaorui Su
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Qiang Yue
- Department of Radiology, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Jianrui Li
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Zhiqiang Zhang
- Department of Diagnostic Radiology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, People's Republic of China
| | - Frederik Barkhof
- UCL Institutes of Neurology and Healthcare Engineering, London, UK
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Raymond Y Huang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ken Chang
- Department of Radiology, Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Haris Sair
- Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chuyang Ye
- School of Information and Electronics, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Zhizheng Zhuo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
6
|
Özdemir İ, Kamson DO, Etyemez S, Blair L, Lin DDM, Barker PB. Downfield Proton MRSI at 3 Tesla: A Pilot Study in Human Brain Tumors. Cancers (Basel) 2023; 15:4311. [PMID: 37686587 PMCID: PMC10486526 DOI: 10.3390/cancers15174311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
PURPOSE To investigate the use of 3D downfield proton magnetic resonance spectroscopic imaging (DF-MRSI) for evaluation of tumor recurrence in patients with glioblastoma (GBM). METHODS Seven patients (4F, age range 44-65 and mean ± standard deviation 59.3 ± 7.5 years) with previously treated GBM were scanned using a recently developed 3D DF-MRSI sequence at 3T. Short TE 3D DF-MRSI and water reference 3D-MRSI scans were collected with a nominal spatial resolution of 0.7 cm3. DF volume data in eight slices covered 12 cm of brain in the cranio-caudal axis. Data were analyzed using the 'LCModel' program and a basis set containing nine peaks ranging in frequency between 6.83 to 8.49 ppm. The DF8.18 (assigned to amides) and DF7.90 peaks were selected for the creation of metabolic images and statistical analysis. Longitudinal MR images and clinical history were used to classify brain lesions as either recurrent tumor or treatment effect, which may include necrosis. DF-MRSI data were compared between lesion groups (recurrent tumor, treatment effect) and normal-appearing brain. RESULTS Of the seven brain tumor patients, two were classified as having recurrent tumor and the rest were classified as treatment effect. Amide metabolite levels from recurrent tumor regions were significantly (p < 0.05) higher compared to both normal-appearing brain and treatment effect regions. Amide levels in lesion voxels classified as treatment effect were significantly lower than normal brain. CONCLUSIONS 3D DF-MRSI in human brain tumors at 3T is feasible and was well tolerated by all patients enrolled in this preliminary study. Amide levels measured by 3D DF-MRSI were significantly different between treatment effect and tumor regrowth.
Collapse
Affiliation(s)
- İpek Özdemir
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - David O. Kamson
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Semra Etyemez
- Department of Obstetrics & Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Lindsay Blair
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Doris D. M. Lin
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Peter B. Barker
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Center for Functional Brain MRI, The Kennedy Krieger Institute, Baltimore, MD 21205, USA
| |
Collapse
|
7
|
Resilience as a Factor Influencing Psychological Distress Experience in Patients with Neuro-Oncological Disease. Curr Oncol 2022; 29:9875-9883. [PMID: 36547190 PMCID: PMC9776769 DOI: 10.3390/curroncol29120776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/05/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer causes psychological distress. Approximately one-third of all patients with cancer suffer from distress requiring psycho-oncological treatment. Examining factors contributing to their distress can inform approaches to counteracting them. Among such factors, resilience is considered to be a psychological adaptive capacity resulting from complex genetic, epigenetic, psychological, and environmental influences. For that reason, we investigated resilience as a factor of psychological distress experience among patients with neuro-oncological disease. To assess distress among patients with neuro-oncological diseases, we performed electronic psycho-oncological screening in the Department of Neurosurgery at Tübingen University Hospital (n = 100) following tumor surgery (T0) using the Resilience Scale 13, the Hornheider Screening Instrument, the Patient Health Questionnaire-2, the Generalized Anxiety Disorder Scale-2, and the Distress Thermometer, all administered on tablets. Follow-up was done 6 months after (T1). The distress of patients with neuro-oncological disease decreased significantly after 6 months (p < 0.01). Most patients (87%) showed moderate to high resilience. Although significant correlations with distress are measurable at the T0 time point (ρ = -0.318 **, p < 0.01), no significant correlations were observed at T1. Thus, resilience seems to significantly impact distress in the acute phase of the neuro-oncological disease. For clinical practice, our findings suggest that resilience-focused screening can provide useful information about patients at risk of experiencing distress.
Collapse
|
8
|
Neuro-Ophthalmic Manifestations of Intracranial Space Occupying Lesions in Adults. BEYOGLU EYE JOURNAL 2022; 7:304-312. [PMID: 36628086 PMCID: PMC9794506 DOI: 10.14744/bej.2022.50469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 01/13/2023]
Abstract
Objectives The purpose of this study is to evaluate the epidemiology, neuro-ophthalmic, and clinical characteristics of intracranial space occupying lesions (ICSOLs) in adult patients. Methods All patients above 16 years presenting with brain tumors confirmed by magnetic resonance imaging and treated surgically in our institute were included in this study. Epidemiology of the patients along with neurological and ophthalmic manifestation was evaluated. Results A total of 252 patients were included in the study ranging from 18 years to 79 years. Supratentorial location was more common than infratentorial location. The most common neurological symptom in our study was headache followed by seizures. Ophthalmic manifestations were present in (73.4%) of patients. The most common visual symptoms and signs were visual loss, strabismus, papilledema, and visual field defects. The most common histopathological diagnosis seen in our study was meningiomas followed by high-grade gliomas. Conclusion Ocular signs and symptoms can be considered as a window to the brain through which ICSOLs can be detected. The most common neurological manifestation of ICSOL in our study was headache with or without true localizing signs and symptoms. More frequently, these patients present to an ophthalmologist before a neurosurgeon with related ocular manifestations. Hence, through our study, we emphasize the importance of a detailed ophthalmological examination in these patients which can aid in early diagnosis and prompt management of such lesions.
Collapse
|
9
|
Follow-Up of High-Grade Glial Tumor; Differentiation of Posttreatment Enhancement and Tumoral Enhancement by DCE-MR Perfusion. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:6948422. [PMID: 35185410 PMCID: PMC8825574 DOI: 10.1155/2022/6948422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/21/2021] [Accepted: 01/11/2022] [Indexed: 02/04/2023]
Abstract
Purpose To search for the utility of DCE-MRP to differentiate between posttreatment enhancement (PT) and tumoral enhancement (TM) in high-grade glial tumors. Materials and Methods Thirty-four patients with glioma (11 grade 3; 23 grade 4) were enrolled. Enhancement in the vicinity of the resection cavity demonstrated by DCE-MRP was taken into consideration. Based on the follow-up scans, reoperation or biopsy results, the enhancement type was categorized as PT or TM. Measurements were performed at the enhancing area near the resection cavity (ERC), nearby (NNA) and contralateral nonenhancing areas (CLNA). Perfusion parameters of the ERC were also subtracted from NNA and CLNA. Intragroup comparison (paired sample t-test) and intergroup comparison (Student's t-test) were made. Results There were 7 PTs and 27 TMs. In the PT, the subtracted values of Ve and IAUC from the CLNA and NNA and the subtracted value of Kep from NNA were statistically different. In TM, all metrics were significantly different comparing the CLNA and NNA. Comparing PT with TM, Ktrans, IAUC, Kep, and subtracted values of Ktrans and IAUC from both NNA and CLNA were significantly different. Conclusions In PT, only Ktrans values did not reveal any difference comparing NNA and CLNA. To differentiate PT from TM, Ktrans, Kep, IAUC, and subtracted values of Ktrans and IAUC from NNA and CLNA can be used. These findings are in concordance with literature.
Collapse
|
10
|
Coppola G, Morris J, Gutierrez-Quintana R, Burnside S, José-López R. Comparison of response assessment in veterinary neuro-oncology and two volumetric neuroimaging methods to assess therapeutic brain tumour responses in veterinary patients. Vet Comp Oncol 2021; 20:404-415. [PMID: 34792828 DOI: 10.1111/vco.12786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/11/2021] [Indexed: 12/13/2022]
Abstract
Standardized veterinary neuroimaging response assessment methods for brain tumours are lacking. Consequently, a response assessment in veterinary neuro-oncology (RAVNO) system which uses the sum product of orthogonal lesion diameters on 1-image section with the largest tumour area, has recently been proposed. In this retrospective study, 22 pre-treatment magnetic resonance imaging (MRI) studies from 18 dogs and four cats with suspected intracranial neoplasia were compared by a single observer to 32 post-treatment MRIs using the RAVNO system and two volumetric methods based on tumour margin or area delineation with HOROS and 3D Slicer software, respectively. Intra-observer variability was low, with no statistically significant differences in agreement index between methods (mean AI ± SD, 0.91 ± 0.06 for RAVNO; 0.86 ± 0.08 for HOROS; and 0.91 ± 0.05 for 3D slicer), indicating good reproducibility. Response assessments consisting of complete or partial responses, and stable or progressive disease, agreed in 23 out of 32 (72%) MRI evaluations using the three methods. The RAVNO system failed to identify changes in mass burden detected with volumetric methods in six cases. 3D Slicer differed from the other two methods in three cases involving cysts or necrotic tissue as it allowed for more accurate exclusion of these structures. The volumetric response assessment methods were more precise in determining changes in absolute tumour burden than RAVNO but were more time-consuming to use. Based on observed agreement between methods, low intra-observer variability and decreased time constraint, RAVNO might be a suitable response assessment method for the clinical setting.
Collapse
Affiliation(s)
- Giovanni Coppola
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Joanna Morris
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Rodrigo Gutierrez-Quintana
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Shona Burnside
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Roberto José-López
- School of Veterinary Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
11
|
Raghavapudi H, Singroul P, Kohila V. Brain Tumor Causes, Symptoms, Diagnosis and Radiotherapy Treatment. Curr Med Imaging 2021; 17:931-942. [PMID: 33573575 DOI: 10.2174/1573405617666210126160206] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 11/22/2022]
Abstract
The strategy used for the treatment of given brain cancer is critical in determining the post effects and survival. An oncological diagnosis of tumor evaluates a range of parameters such as shape, size, volume, location and neurological complexity that define the symptomatic severity. The evaluation determines a suitable treatment approach chosen from a range of options such as surgery, chemotherapy, hormone therapy, radiation therapy and other targeted therapies. Often, a combination of such therapies is applied to achieve superior results. Radiotherapy serves as a better treatment strategy because of a higher survival rate. It offers the flexibility of synergy with other treatment strategies and fewer side effects on organs at risk. This review presents a radiobiological perspective in the treatment of brain tumor. The cause, symptoms, diagnosis, treatment, post-treatment effects and the framework involved in its elimination are summarized.
Collapse
Affiliation(s)
- Haarika Raghavapudi
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| | - Pankaj Singroul
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| | - V Kohila
- Department of Biotechnology, National Institute of Technology Warangal, Warangal -506004, Telangana, India
| |
Collapse
|
12
|
Nguyen P, Doan P, Rimpilainen T, Konda Mani S, Murugesan A, Yli-Harja O, Candeias NR, Kandhavelu M. Synthesis and Preclinical Validation of Novel Indole Derivatives as a GPR17 Agonist for Glioblastoma Treatment. J Med Chem 2021; 64:10908-10918. [PMID: 34304559 PMCID: PMC8389915 DOI: 10.1021/acs.jmedchem.1c00277] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The discovery of a potential ligand-targeting G protein-coupled receptor 17 (GPR17) is important for developing chemotherapeutic agents against glioblastoma multiforme (GBM). We used the integration of ligand- and structure-based cheminformatics and experimental approaches for identifying the potential GPR17 ligand for GBM treatment. Here, we identified a novel indoline-derived phenolic Mannich base as an activator of GPR17 using molecular docking of over 6000 indoline derivatives. One of the top 10 hit molecules, CHBC, with a glide score of -8.390 was synthesized through a multicomponent Petasis borono-Mannich reaction. The CHBC-GPR17 interaction leads to a rapid decrease of cAMP and Ca2+. CHBC exhibits the cytotoxicity effect on GBM cells in a dose-dependent manner with an IC50 of 85 μM, whereas the known agonist MDL29,951 showed a negligible effect. Our findings suggest that the phenolic Mannich base could be a better GPR17 agonist than MDL29,951, and further uncovering their pharmacological properties could potentiate an inventive GBM treatment.
Collapse
Affiliation(s)
- Phung Nguyen
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, 33720 Tampere, Finland.,BioMeditech and Tays Cancer Center, Tampere University, Hospital, P.O. Box 553, 33101 Tampere, Finland
| | - Phuong Doan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, 33720 Tampere, Finland.,BioMeditech and Tays Cancer Center, Tampere University, Hospital, P.O. Box 553, 33101 Tampere, Finland
| | - Tatu Rimpilainen
- Faculty of Engineering and Natural Sciences, Tampere University, 33101 Tampere, Finland
| | - Saravanan Konda Mani
- Scigen Research and Innovation Pvt Ltd, Periyar Technology Business Incubator, Thanjavur, Tamil Nadu 613403, India
| | - Akshaya Murugesan
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, 33720 Tampere, Finland.,Department of Biotechnology, Lady Doak College, Thallakulam, 625002 Madurai, India
| | - Olli Yli-Harja
- Computational Systems Biology Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland.,Institute for Systems Biology, 1441N 34th Street, Seattle, Washington 98103-8904, United States
| | - Nuno R Candeias
- Faculty of Engineering and Natural Sciences, Tampere University, 33101 Tampere, Finland.,LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Meenakshisundaram Kandhavelu
- Molecular Signaling Lab, Faculty of Medicine and Health Technology, Tampere University, 33720 Tampere, Finland.,BioMeditech and Tays Cancer Center, Tampere University, Hospital, P.O. Box 553, 33101 Tampere, Finland
| |
Collapse
|
13
|
Miles X, Vandevoorde C, Hunter A, Bolcaen J. MDM2/X Inhibitors as Radiosensitizers for Glioblastoma Targeted Therapy. Front Oncol 2021; 11:703442. [PMID: 34307171 PMCID: PMC8296304 DOI: 10.3389/fonc.2021.703442] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Inhibition of the MDM2/X-p53 interaction is recognized as a potential anti-cancer strategy, including the treatment of glioblastoma (GB). In response to cellular stressors, such as DNA damage, the tumor suppression protein p53 is activated and responds by mediating cellular damage through DNA repair, cell cycle arrest and apoptosis. Hence, p53 activation plays a central role in cell survival and the effectiveness of cancer therapies. Alterations and reduced activity of p53 occur in 25-30% of primary GB tumors, but this number increases drastically to 60-70% in secondary GB. As a result, reactivating p53 is suggested as a treatment strategy, either by using targeted molecules to convert the mutant p53 back to its wild type form or by using MDM2 and MDMX (also known as MDM4) inhibitors. MDM2 down regulates p53 activity via ubiquitin-dependent degradation and is amplified or overexpressed in 14% of GB cases. Thus, suppression of MDM2 offers an opportunity for urgently needed new therapeutic interventions for GB. Numerous small molecule MDM2 inhibitors are currently undergoing clinical evaluation, either as monotherapy or in combination with chemotherapy and/or other targeted agents. In addition, considering the major role of both p53 and MDM2 in the downstream signaling response to radiation-induced DNA damage, the combination of MDM2 inhibitors with radiation may offer a valuable therapeutic radiosensitizing approach for GB therapy. This review covers the role of MDM2/X in cancer and more specifically in GB, followed by the rationale for the potential radiosensitizing effect of MDM2 inhibition. Finally, the current status of MDM2/X inhibition and p53 activation for the treatment of GB is given.
Collapse
Affiliation(s)
- Xanthene Miles
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Alistair Hunter
- Radiobiology Section, Division of Radiation Oncology, Department of Radiation Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| |
Collapse
|
14
|
ctDNA-Based Liquid Biopsy of Cerebrospinal Fluid in Brain Cancer. Cancers (Basel) 2021; 13:cancers13091989. [PMID: 33919036 PMCID: PMC8122255 DOI: 10.3390/cancers13091989] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/18/2022] Open
Abstract
The correct characterisation of central nervous system (CNS) malignancies is crucial for accurate diagnosis and prognosis and also the identification of actionable genomic alterations that can guide the therapeutic strategy. Surgical biopsies are performed to characterise the tumour; however, these procedures are invasive and are not always feasible for all patients. Moreover, they only provide a static snapshot and can miss tumour heterogeneity. Currently, monitoring of CNS cancer is performed by conventional imaging techniques and, in some cases, cytology analysis of the cerebrospinal fluid (CSF); however, these techniques have limited sensitivity. To overcome these limitations, a liquid biopsy of the CSF can be used to obtain information about the tumour in a less invasive manner. The CSF is a source of cell-free circulating tumour DNA (ctDNA), and the analysis of this biomarker can characterise and monitor brain cancer. Recent studies have shown that ctDNA is more abundant in the CSF than plasma for CNS malignancies and that it can be sequenced to reveal tumour heterogeneity and provide diagnostic and prognostic information. Furthermore, analysis of longitudinal samples can aid patient monitoring by detecting residual disease or even tracking tumour evolution at relapse and, therefore, tailoring the therapeutic strategy. In this review, we provide an overview of the potential clinical applications of the analysis of CSF ctDNA and the challenges that need to be overcome in order to translate research findings into a tool for clinical practice.
Collapse
|
15
|
Multimodal assessment of disease activity in glioblastoma : A single center experience. Wien Klin Wochenschr 2021; 133:1148-1154. [PMID: 33877437 DOI: 10.1007/s00508-021-01859-2] [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: 02/11/2021] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Assessment of disease activity in glioblastoma (GBM) can be challenging due to several clinical and radiological pitfalls. Besides MRI, FET-PET and neurocognitive assessment (NA) are used in several neuro-oncological centers in order to improve the specificity of response assessment. We performed a retrospective study to investigate whether the assessment by RANO (Response Assessment in NeuroOncology) corresponds to FET-PET imaging and NA results. Moreover, the concordance of RANO with a final recommendation of an interdisciplinary neuro-oncological tumor board recommendation (TBR) was analyzed. METHODS We enrolled 25 consecutive patients with newly diagnosed histologically confirmed GBM in a pilot study, accounting for 81 multimodal test results. All patients were selected after undergoing consecutive follow-up comprising MRI, FET-PET, and NA with a subsequent TBR. Results were analyzed for correlations between RANO, FET-PET and NA. An additional consistency analysis was performed to elucidate the impact of RANO on decision making. RESULTS A highly statistically significant correlation was found between RANO and FET-PET and NA results (all P < 0.01); however, 26% of follow-up tests exhibited inconsistent results in multimodal assessment, among which RANO was only 48% in accordance with the final TBR. The concordance of NA and FET-PET with the final TBR was 67% and 86%, respectively. CONCLUSION The RANO proved its value in the context of multimodal assessment of disease activity in GBM; however, because the implementation of multimodal assessment showed a considerably high percentage of inconsistent results, further studies are required to investigate the relationship between different assessment techniques, in addition to their overall significance to response rating.
Collapse
|
16
|
Zhang X, Wei X, Liu J, Yang J, Jin P. Up-regulation of long non-coding RNA BLACAT1 predicts aggressive clinicopathologic characteristics and poor prognosis of glioma. Medicine (Baltimore) 2021; 100:e20722. [PMID: 33725920 PMCID: PMC7982219 DOI: 10.1097/md.0000000000020722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/18/2020] [Accepted: 05/10/2020] [Indexed: 01/05/2023] Open
Abstract
ABSTRACT Bladder cancer-associated transcript 1 (BLACAT1) is one of the most common cancer-associated long non-coding RNAs (lncRNAs), which has been reported as a tumor promotor in several malignancies. Previously, BLACAT1 was found to be overexpressed in glioma tissues and cell lines. Functional assays determined that BLACAT1 promoted glioma cell proliferation, migration, invasion and epithelial-mesenchymal transition, suggesting that BLACAT1 might serve as an oncogene in glioma. In the present study, we aimed to investigate its clinical significance and prognostic value in glioma patients.A total of 137 paired glioma tissue samples and adjacent normal brain tissue samples were collected from 137 glioma patients who underwent surgery from May 2014 to February 2019. The Student t test was applied to determine the statistical significance of the observed differences between 2 groups. Survival curves were constructed and differences among groups were calculated using the Kaplan-Meier method.The relative expression of BLACAT1 in glioma samples was significantly higher than that of matched normal tissues (P < .001). The expression level of tissue BLACAT1 was statistically correlated with tumor size (P = .04), Karnofsky Performance Status (KPS) (P = .006), and WHO grade (P = .017). Kaplan-Meier analysis with the log-rank test revealed that BLACAT1 up-regulation was correlated with shorter overall survival time of patients with glioma (Log Rank test, P = .012). In multivariate Cox analysis, BLACAT1 expression was found to be an independent prognostic factor for overall survival in patients with glioma (HR = 2.739; 95% CI: 1.785-8.229; P = .035). Our study demonstrates that up-regulation of BLACAT1 is able to predict aggressive clinicopathologic characteristics and poor prognosis of glioma patients. These findings may have significant implications for potential treatment options and prognosis for patients with glioma.
Collapse
Affiliation(s)
- Xiaojue Zhang
- Department of Craniocerebral Surgery, People's Hospital of Lanling County, Lanling, Linyi
| | - Xiuchuan Wei
- Department of Craniocerebral Surgery, People's Hospital of Lanling County, Lanling, Linyi
| | - Jie Liu
- Department of Craniocerebral Surgery, People's Hospital of Lanling County, Lanling, Linyi
| | - Jiaying Yang
- Department of Craniocerebral Surgery, People's Hospital of Lanling County, Lanling, Linyi
| | - Peng Jin
- Department of Neurosurgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
17
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Neuroimaging Clin N Am 2021; 31:103-120. [PMID: 33220823 DOI: 10.1016/j.nic.2020.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
18
|
Debelee TG, Kebede SR, Schwenker F, Shewarega ZM. Deep Learning in Selected Cancers' Image Analysis-A Survey. J Imaging 2020; 6:121. [PMID: 34460565 PMCID: PMC8321208 DOI: 10.3390/jimaging6110121] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 02/08/2023] Open
Abstract
Deep learning algorithms have become the first choice as an approach to medical image analysis, face recognition, and emotion recognition. In this survey, several deep-learning-based approaches applied to breast cancer, cervical cancer, brain tumor, colon and lung cancers are studied and reviewed. Deep learning has been applied in almost all of the imaging modalities used for cervical and breast cancers and MRIs for the brain tumor. The result of the review process indicated that deep learning methods have achieved state-of-the-art in tumor detection, segmentation, feature extraction and classification. As presented in this paper, the deep learning approaches were used in three different modes that include training from scratch, transfer learning through freezing some layers of the deep learning network and modifying the architecture to reduce the number of parameters existing in the network. Moreover, the application of deep learning to imaging devices for the detection of various cancer cases has been studied by researchers affiliated to academic and medical institutes in economically developed countries; while, the study has not had much attention in Africa despite the dramatic soar of cancer risks in the continent.
Collapse
Affiliation(s)
- Taye Girma Debelee
- Artificial Intelligence Center, 40782 Addis Ababa, Ethiopia; (S.R.K.); (Z.M.S.)
- College of Electrical and Mechanical Engineering, Addis Ababa Science and Technology University, 120611 Addis Ababa, Ethiopia
| | - Samuel Rahimeto Kebede
- Artificial Intelligence Center, 40782 Addis Ababa, Ethiopia; (S.R.K.); (Z.M.S.)
- Department of Electrical and Computer Engineering, Debreberhan University, 445 Debre Berhan, Ethiopia
| | - Friedhelm Schwenker
- Institute of Neural Information Processing, University of Ulm, 89081 Ulm, Germany;
| | | |
Collapse
|
19
|
Nia HT, Datta M, Seano G, Zhang S, Ho WW, Roberge S, Huang P, Munn LL, Jain RK. In vivo compression and imaging in mouse brain to measure the effects of solid stress. Nat Protoc 2020; 15:2321-2340. [PMID: 32681151 PMCID: PMC11412114 DOI: 10.1038/s41596-020-0328-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/06/2020] [Indexed: 02/07/2023]
Abstract
We recently developed an in vivo compression device that simulates the solid mechanical forces exerted by a growing tumor on the surrounding brain tissue and delineates the physical versus biological effects of a tumor. This device, to our knowledge the first of its kind, can recapitulate the compressive forces on the cerebellar cortex from primary (e.g., glioblastoma) and metastatic (e.g., breast cancer) tumors, as well as on the cerebellum from tumors such as medulloblastoma and ependymoma. We adapted standard transparent cranial windows normally used for intravital imaging studies in mice to include a turnable screw for controlled compression (acute or chronic) and decompression of the cerebral cortex. The device enables longitudinal imaging of the compressed brain tissue over several weeks or months as the screw is progressively extended against the brain tissue to recapitulate tumor growth-induced solid stress. The cranial window can be simply installed on the mouse skull according to previously established methods, and the screw mechanism can be readily manufactured in-house. The total time for construction and implantation of the in vivo compressive cranial window is <1 h (per mouse). This technique can also be used to study a variety of other diseases or disorders that present with abnormal solid masses in the brain, including cysts and benign growths.
Collapse
Affiliation(s)
- Hadi T Nia
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Meenal Datta
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Giorgio Seano
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Tumor Microenvironment Laboratory, Institut Curie Research Center, Paris-Saclay University, PSL Research University, Inserm U1021, CNRS UMR3347, Orsay, France
| | - Sue Zhang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - William W Ho
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sylvie Roberge
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Peigen Huang
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
20
|
Ge BH, Li GC. Long non-coding RNA SNHG17 promotes proliferation, migration and invasion of glioma cells by regulating the miR-23b-3p/ZHX1 axis. J Gene Med 2020; 22:e3247. [PMID: 32602607 DOI: 10.1002/jgm.3247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Long non-coding RNA (lncRNA) small nucleolar RNA host gene 17 (SNHG17) is a carcinogenic lncRNA in diverse cancers. The expression pattern and mechanisms of SNHG17 in glioma still await verification. METHODS Paired glioma samples were enrolled. SNHG17, miR-23b-3p, and zinc-fingers and homeoboxes 1 (ZHX1) mRNA expression were examined by a quantitative real-time polymerase chain reaction (qRT-PCR). SNHG17 short hairpin RNA (shRNA) and miR-23b-3p mimics were transfected into LN229 and U251 cell lines to repress SNHG17 and up-regulate miR-23b-3p expression, respectively. Proliferation, migration and invasion of LN229 and U251 cells were probed by a cell counting kit-8 assay and a Transwell assay. Bioinformatics prediction, dual-luciferase reporter assay, RNA immunoprecipitation assay, qRT-PCR and western blotting were applied to determine the regulatory relationships among SNHG17, miR-23b-3p and ZHX1. RESULTS SNHG17 expression was markedly raised in glioma tissues, which was positively correlated with ZHX1 expression and negatively associated with the expression of miR-23b-3p. After transfection of SNHG17 shRNAs into glioma cells, the proliferation, migration and invasion of cancer cells was markedly restrained. miR-23b-3p mimics the function of SHNG17 knockdown. Furthermore, miR-23b-3p was shown to be negatively modulated by SNHG17, and ZHX1 was identified as a target of miR-23b-3p. CONCLUSIONS SNHG17 is a "competing endogenous RNA" with respect to modulating ZHX1 expression by adsorbing miR-23b-3p and thereby promoting glioma progression.
Collapse
Affiliation(s)
- Bei-Hai Ge
- Department of Neurology, Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou, Guangxi, China
| | - Guo-Cheng Li
- Department of Neurosurgery, Guangxi Zhuang Autonomous Region Brain Hospital, Liuzhou, Guangxi, China
| |
Collapse
|
21
|
Liu X, Huang H, Li X, Zheng X, Zhou C, Xue B, He J, Zhang Y, Liu L. Knockdown of ADAMDEC1 inhibits the progression of glioma in vitro. Histol Histopathol 2020; 35:997-1005. [PMID: 32378728 DOI: 10.14670/hh-18-227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Glioma is one of the most lethal malignant tumors all over the world. The prognosis of patients with high‑grade glioma remains very poor. Therefore, it is urgent to find a novel strategy for the treatment of glioma. It has been reported that ADAMDEC1 could regulate the progression of multiple diseases, including cancers. However, the role of ADAMDEC1 during the tumorigenesis of glioma remains largely unknown. Methods, Gene expression of ADAMDEC1 in glioma tissues or in cells was detected by qRT-PCR. Western blot was performed to measure the protein expressions of p53, active caspase3, active caspase9, CDK2 and Cyclin A in glioma cells. Cell proliferation was detected by CCK-8 assay. Cell apoptosis or cycle was tested by flow cytometry. Transwell was used to test the invasion of glioma cells. RESULTS The expression of ADAMDEC1 in glioma tissues or cells was significantly upregulated. In addition, downregulation of ADAMDEC1 notably inhibited the proliferation and induced apoptosis of glioma cells through upregulation of active caspase 3 and active caspase 9. Besides, silencing of ADAMDEC1 obviously induced G1 arrest in glioma cells via modulation of cell cycle-related proteins. Finally, knockdown of ADAMDEC1 significantly inhibited the migration and invasion of glioma cells. In contrast, overexpression of ADAMDEC1 promoted cell proliferation, migration and invasion of glioma cells. CONCLUSION Downregulation of ADAMDEC1 could significantly inhibit the tumorigenesis of glioma in vitro, which may serve as a novel target for the treatment of glioma.
Collapse
Affiliation(s)
- Xueliang Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Huang
- Department of Neurosurgery, Guang'an People's Hospital, Guangan, Sichuan, China
| | - Xuehan Li
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaomei Zheng
- Department of Neurology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Chong Zhou
- Department of Neurology, Jianyang People's Hospital, Jianyang, Sichuan, China
| | - Bin Xue
- Department of Neurosurgery, Nanbu People's Hospital, Nanchong, Sichuan, China
| | - Jimin He
- Department of Neurosurgery, Suining Central Hospital, Suining, Sichuan, China
| | - Ye Zhang
- Department of Neurosurgery, Mianyang Central Hospital, Mianyang, Sichuan, China
| | - Liang Liu
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
22
|
Eichberg DG, Di L, Morell AA, Shah AH, Semonche AM, Chin CN, Bhatia RG, Jamshidi AM, Luther EM, Komotar RJ, Ivan ME. Incidence of high grade gliomas presenting as radiographically non-enhancing lesions: experience in 111 surgically treated non-enhancing gliomas with tissue diagnosis. J Neurooncol 2020; 147:671-679. [PMID: 32221785 DOI: 10.1007/s11060-020-03474-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/23/2020] [Indexed: 02/03/2023]
Abstract
PURPOSE Although non-enhancing lesions suspicious for glioma are usually assumed to be low grade glioma (LGG), some high grade glioma (HGG) do not enhance, which may lead to a delay in biopsy and/or resection, diagnosis, and treatment initiation. Thus, there is a clear need for a large-sample study that quantifies the rate of malignant, non-enhancing gliomas. METHODS We retrospectively reviewed our series of 561 consecutive surgically treated gliomas with tissue diagnosis, 111 of which were non-enhancing, to determine the prevalence of high-grade histology in radiographically presumed LGG. Relative expression of tumor markers were also reported for non-enhancing lesions to investigate genetic correlates. RESULTS We identified 561 surgically treated gliomas with tissue diagnosis from August 2012 to July 2018 and found that 111 patients (19.8%) demonstrated non-enhancing lesions suspicious for glioma on preoperative MRI. Thirty-one (27.9%) of the non-enhancing lesions were classified as HGGs (WHO Grade III or IV). Non-enhancing lesions were four times more likely to be HGG in patients older than 60 years than patients younger than 35 years (41.2% vs. 11.4%, Pearson Chi2 p < 0.001). Binomial logistic regression showed a significant inverse effect of age on the presence of IDH mutation in non-enhancing HGGs (p = 0.007). CONCLUSION A clinically significant proportion (27.9%) of non-enhancing lesions were found to be HGG on final pathologic diagnosis. Thus, in patients with good functional and health status, especially those older than 60 years, we recommend obtaining tissue diagnosis of all lesions suspected to be glioma, even those that are non-enhancing, to guide diagnosis as well as early initiation of chemotherapy and radiation therapy.
Collapse
Affiliation(s)
- Daniel G Eichberg
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA.
| | - Long Di
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Alexis A Morell
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Ashish H Shah
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Alexa M Semonche
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Christopher N Chin
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Rita G Bhatia
- Department of Radiology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aria M Jamshidi
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Evan M Luther
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
| | - Ricardo J Komotar
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Michael E Ivan
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Lois Pope Life Center, 1095 NW 14th Terrace (D4-6), Miami, FL, 33146, USA
- Sylvester Comprehensive Cancer Center, Miami, FL, USA
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW H3K27M is a frequent histone mutation within diffuse midline gliomas and is associated with a dismal prognosis, so much so that the 2016 CNS WHO classification system created a specific category of "Diffuse Midline Glioma, H3K27M-mutant". Here we outline the latest pre-clinical data and ongoing current clinical trials that target H3K27M, as well as explore diagnosis and treatment monitoring by serial liquid biopsy. RECENT FINDINGS Multiple epigenetic compounds have demonstrated efficacy and on-target effects in pre-clinical models. The imipridone ONC201 and the IDO1 inhibitor indoximod have demonstrated early clinical activity against H3K27M-mutant gliomas. Liquid biopsy of cerebrospinal fluid has shown promise for clinical use in H3K27M-mutant tumors for diagnosis and monitoring treatment response. While H3K27M has elicited a widespread platform of pre-clinical therapies with promise, much progress still needs to be made to improve outcomes for diffuse midline glioma patients. We present current treatment and monitoring techniques as well as novel approaches in identifying and targeting H3K27M-mutant gliomas.
Collapse
|
24
|
Verhoeven J, Baguet T, Piron S, Pauwelyn G, Bouckaert C, Descamps B, Raedt R, Vanhove C, De Vos F, Goethals I. 2-[ 18F]FELP, a novel LAT1-specific PET tracer, for the discrimination between glioblastoma, radiation necrosis and inflammation. Nucl Med Biol 2019; 82-83:9-16. [PMID: 31841816 DOI: 10.1016/j.nucmedbio.2019.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Considering the need for rapid change of treatment in recurrent glioblastoma (GB), it is of utmost importance to characterize PET radiopharmaceuticals that allow early discrimination of tumor from therapy-related effects. In this study, we examined the value of 2-[18F]FELP as a LAT1 tumor-specific PET tracer in comparison with [18F]FDG and [18F]FET in a combined orthotopic rat radiation necrosis and glioblastoma model. A second experiment compared 2-[18F]FELP to [18F]FDG in a mouse glioblastoma - inflammation model. METHODS Using the small animal radiation research platform (SARRP), radiation necrosis (RN) was induced in the left frontal lobe of the rat brain. When radiation-induced changes were visible on MRI, F98 rat glioblastoma cells were stereotactically inoculated in the contralateral right frontal lobe. When tumor growth was confirmed on MRI, 2-[18F]FELP, [18F]FET and [18F]FDG PET scans were acquired on three consecutive days. In an inflammation experiment, mice were inoculated in the left thigh with U87 human glioblastoma cells. After heterotopic tumor growth was confirmed macroscopically, inflammation was induced by injection of turpentine subcutaneously in the right thigh. Subsequently, 2-[18F]FELP and [18F]FDG scans were acquired on two consecutive days. RESULTS The in vivo PET images demonstrated that 2-[18F]FELP could differentiate glioblastoma and radiation necrosis using SUVmean (p = 0.0016) and LNRmean (p = 0.009), while [18F]FET was only able to differentiate both lesions by means of the SUVmean. (p = 0.047) Delayed [18F]FDGlate PET (4 h postinjection) was also able to distinguish glioblastoma from radiation necrosis, but smaller lesion-to-normal brain ratios were observed (SUVmean: p = 0.009; LNRmean: p = 0.028). In the inflammation study, 2-[18F]FELP showed no significant uptake in the inflammation lesion when compared to the control group (SUVmean: p = 0.149; LNRmean: p = 0.083). In contrast, both conventional and delayed [18F]FDG displayed significant uptake in the turpentine-invoked lesion (SUVmean: p = 0.021; LNRmean: p = 0.021). CONCLUSION This study suggests that the 2-[18F]FELP PET is able to differentiate glioblastoma from radiation necrosis and that the 2-[18F]FELP uptake is less likely to be contaminated by the presence of inflammation than the [18F]FDG signal. ADVANCES IN KNOWLEDGE These results are clinically relevant for the differential diagnosis between tumor and radiation necrosis because radiation necrosis always contains a certain amount of inflammatory cells. Hence, 2-[18F]FELP is preferred to discriminate tumor from radiation necrosis.
Collapse
Affiliation(s)
| | - Tristan Baguet
- Laboratory for Radiopharmacy, Ghent University, Ghent, Belgium
| | - Sarah Piron
- Laboratory for Radiopharmacy, Ghent University, Ghent, Belgium
| | - Glenn Pauwelyn
- Laboratory for Radiopharmacy, Ghent University, Ghent, Belgium
| | - Charlotte Bouckaert
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Robrecht Raedt
- Laboratory for Clinical and Experimental Neurophysiology, Neurobiology and Neuropsychology (LCEN3), Ghent University Hospital, Ghent, Belgium
| | - Christian Vanhove
- IBiTech-MEDISIP, Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Filip De Vos
- Laboratory for Radiopharmacy, Ghent University, Ghent, Belgium
| | - Ingeborg Goethals
- Department of Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
25
|
Chen H, Li Q, Liang J, Jin M, Lu A. LncRNA CPS1-IT1 serves as anti-oncogenic role in glioma. Biomed Pharmacother 2019; 118:109277. [PMID: 31545272 DOI: 10.1016/j.biopha.2019.109277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/24/2019] [Accepted: 07/25/2019] [Indexed: 01/16/2023] Open
Abstract
Carbamoyl-phosphate synthetase 1 intronic transcript 1 (CPS1-IT1) is identified recently as a novel tumor suppressive long non-coding RNA (lncRNA). However, the expression status and clinical significance of CPS1-IT1 expression remained unknown in glioma. In our study, we observed CPS1-IT1 levels were decreased in glioma tissues and cells compared with paired normal brain tissues and human astrocyte cell line, respectively. Moreover, we analyzed the associations of CPS1-IT1 expression with clinicopathological characteristics, and found low CPS1-IT1 expression was correlated with high World Health Organisation (WHO) grade and large tumor size in glioma patients. Survival analysis showed glioma patients in low CPS1-IT1 expression group had shorter overall survival than those in high CPS1-IT1 expression group, and low CPS1-IT1 expression was an independent prognostic factor for overall survival in glioma patients. The in vitro studies suggested up-regulation of CPS1-IT1 expression resulted in the decrease of proliferation, migration and invasion abilities of glioma cells. In conclusion, CPS1-IT1 plays an anti-oncogenic role in glioma.
Collapse
Affiliation(s)
- Hengsan Chen
- Department of Neurosurgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China.
| | - Qiang Li
- Department of Neurosurgery, Second Affiliated Hospital of Lanzhou University, Lanzhou, Gansu 730030, China
| | - Jin Liang
- Department of Neurosurgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Ming Jin
- Department of Neurosurgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China
| | - Anqing Lu
- Department of Neurosurgery, Gansu Provincial Hospital, Lanzhou, Gansu 730000, China.
| |
Collapse
|
26
|
Strauss SB, Meng A, Ebani EJ, Chiang GC. Imaging Glioblastoma Posttreatment: Progression, Pseudoprogression, Pseudoresponse, Radiation Necrosis. Radiol Clin North Am 2019; 57:1199-1216. [PMID: 31582045 DOI: 10.1016/j.rcl.2019.07.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Radiographic monitoring of posttreatment glioblastoma is important for clinical trials and determining next steps in management. Evaluation for tumor progression is confounded by the presence of treatment-related radiographic changes, making a definitive determination less straight-forward. The purpose of this article was to describe imaging tools available for assessing treatment response in glioblastoma, as well as to highlight the definitions, pathophysiology, and imaging features typical of true progression, pseudoprogression, pseudoresponse, and radiation necrosis.
Collapse
Affiliation(s)
- Sara B Strauss
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Alicia Meng
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Edward J Ebani
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA
| | - Gloria C Chiang
- Department of Radiology, Weill Cornell Medical Center, 525 East 68th Street, Box 141, New York, NY 10065, USA.
| |
Collapse
|
27
|
Fiorentini G, Sarti D, Milandri C, Dentico P, Mambrini A, Fiorentini C, Mattioli G, Casadei V, Guadagni S. Modulated Electrohyperthermia in Integrative Cancer Treatment for Relapsed Malignant Glioblastoma and Astrocytoma: Retrospective Multicenter Controlled Study. Integr Cancer Ther 2019; 18:1534735418812691. [PMID: 30580645 PMCID: PMC7240877 DOI: 10.1177/1534735418812691] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/09/2018] [Accepted: 10/12/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND There are interesting studies on glioma therapy with modulated electrohyperthermia (mEHT), which combines heat therapy with an electric field. Clinical researchers not only found the mEHT method feasible for palliation but also reported evidence of therapeutic response. PURPOSE To study the efficacy and safety of mEHT for the treatment of relapsed malignant glioma and astrocytoma versus best supportive care (BSC). METHODS We collected data retrospectively on 149 patients affected by malignant glioma and astrocytoma. Inclusion criteria were informed consent signed; >18 years old; histological diagnosis of malignant glioma or astrocytoma; relapsed after surgery, adjuvant temozolomide-based chemotherapy, and radiotherapy; and indication for treatment with mEHT in palliative setting. mEHT was performed with capacitive coupling technique keeping the skin surface at 26°C and the tumor temperature at 40°C to 42.5°C for > 90% of treatment duration (20-60 minutes). The applied power was 40 to 150 W using a step-up heating protocol. Results from patients treated with mEHT were compared with those treated with BSC. RESULTS A total of 149 consecutive patients were enrolled in the study, 111 (74%) had glioblastoma multiforme (GBM), and 38 (26%) had astrocytoma (AST). mEHT was performed for 28 (25%) of GBM and 24 (63%) of AST patients. Tumor response at the 3-month follow-up was observed in 29% and 48% of GBM and AST patients after mEHT, and in 4% and 10% of GBM and AST patients after BSC, respectively. The survival rate at first and second year in the mEHT group was 77.3% and 40.9% for AST, and 61% and 29% for GBM, respectively. The 5-year overall survival of AST was 83% after mEHT versus 25% after BSC and 3.5% after mEHT versus 1.2% after BSC for GBM. The median overall survival of mEHT was 14 months (range 2-108 months) for GBM and 16.5 months (range 3-156 months) for the AST group. We observed 4 long-term survivors in the AST and 2 in the GBM group. Two of the long survivors in AST and 1 in GBM group were treated by mEHT. CONCLUSIONS mEHT in integrative therapy may have a promising role in the treatment and palliation of relapsed GBM and AST.
Collapse
Affiliation(s)
| | - Donatella Sarti
- Azienda Ospedaliera “Ospedali Riuniti
Marche Nord”, Pesaro, Italy
| | - Carlo Milandri
- Nuovo Ospedale San Giuseppe, ASL Toscana
Centro, Empoli, Florence, Italy
| | - Patrizia Dentico
- Nuovo Ospedale San Giuseppe, ASL Toscana
Centro, Empoli, Florence, Italy
| | | | | | | | - Virginia Casadei
- Azienda Ospedaliera “Ospedali Riuniti
Marche Nord”, Pesaro, Italy
| | | |
Collapse
|
28
|
Khan UA, Rennert RC, White NS, Bartsch H, Farid N, Dale AM, Chen CC. Diagnostic utility of restriction spectrum imaging (RSI) in glioblastoma patients after concurrent radiation-temozolomide treatment: A pilot study. J Clin Neurosci 2018; 58:136-141. [PMID: 30253908 DOI: 10.1016/j.jocn.2018.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 05/01/2018] [Accepted: 09/10/2018] [Indexed: 01/21/2023]
Abstract
Discriminating between tumor recurrence and treatment effects in glioblastoma patients undergoing radiation-temozolomide (RT/TMZ) therapy remains a major clinical challenge. Here, we report a pilot study to determine the utility of restriction spectrum imaging (RSI), an advanced diffusion-weighted MRI (DWI) technique that affords meso-scale resolution of cell density, in this assessment. A retrospective review of 31 patients with glioblastoma treated between 2011 and 2017 who underwent surgical resection or biopsy over radiographic concern for tumor recurrence following RT/TMZ was performed. All patients underwent RSI prior to surgical resection. Diagnostic utility of RSI for tumor recurrence was determined in comparison to histopathology. Analysis of surgical specimens revealed treatment effects in 6/31 patients (19%) and tumor recurrence in 25/31 patients (81%). There was general concordance between the measured RSI signal and histopathologic diagnosis. RSI was negative in 5/6 patients (83%) in patients with histological evidence of treatment effects. RSI was positive in 21/25 patients (84%) in patients with tumor recurrence. The sensitivity, specificity, positive and negative predictive values of RSI for glioblastoma recurrence were 84%, 86%, 95%, and 60%, respectively. Histopathologic review showed agreement between the RSI signal and cellularity of the tumor specimen. These data support the use of RSI in the evaluation of treatment effects versus tumor recurrence in glioblastoma patients after RT-TMZ therapy.
Collapse
Affiliation(s)
- Usman A Khan
- Department of Neurosurgery, University of California San Diego, San Diego, CA, USA
| | - Robert C Rennert
- Department of Neurosurgery, University of California San Diego, San Diego, CA, USA
| | - Nathan S White
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Hauke Bartsch
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Nikdokht Farid
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA; Department of Radiology, University of California San Diego, La Jolla, CA, USA
| | - Anders M Dale
- Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
29
|
Boult JKR, Box G, Vinci M, Perryman L, Eccles SA, Jones C, Robinson SP. Evaluation of the Response of Intracranial Xenografts to VEGF Signaling Inhibition Using Multiparametric MRI. Neoplasia 2017; 19:684-694. [PMID: 28780387 PMCID: PMC5547238 DOI: 10.1016/j.neo.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/15/2017] [Accepted: 05/15/2017] [Indexed: 11/29/2022] Open
Abstract
Vascular endothelial growth factor A (VEGF-A) is considered one of the most important factors in tumor angiogenesis, and consequently, a number of therapeutics have been developed to inhibit VEGF signaling. Therapeutic strategies to target brain malignancies, both primary brain tumors, particularly in pediatric patients, and metastases, are lacking, but targeting angiogenesis may be a promising approach. Multiparametric MRI was used to investigate the response of orthotopic SF188luc pediatric glioblastoma xenografts to small molecule pan-VEGFR inhibitor cediranib and the effects of both cediranib and cross-reactive human/mouse anti-VEGF-A antibody B20-4.1.1 in intracranial MDA-MB-231 LM2–4 breast cancer xenografts over 48 hours. All therapeutic regimens resulted in significant tumor growth delay. In cediranib-treated SF188luc tumors, this was associated with lower Ktrans (compound biomarker of perfusion and vascular permeability) than in vehicle-treated controls. Cediranib also induced significant reductions in both Ktrans and apparent diffusion coefficient (ADC) in MDA-MB-231 LM2–4 tumors associated with decreased histologically assessed perfusion. B20-4.1.1 treatment resulted in decreased Ktrans, but in the absence of a change in perfusion; a non-significant reduction in vascular permeability, assessed by Evans blue extravasation, was observed in treated tumors. The imaging responses of intracranial MDA-MB-231 LM2–4 tumors to VEGF/VEGFR pathway inhibitors with differing mechanisms of action are subtly different. We show that VEGF pathway blockade resulted in tumor growth retardation and inhibition of tumor vasculature in preclinical models of pediatric glioblastoma and breast cancer brain metastases, suggesting that multiparametric MRI can provide a powerful adjunct to accelerate the development of antiangiogenic therapies for use in these patient populations.
Collapse
Affiliation(s)
- Jessica K R Boult
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Gary Box
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Maria Vinci
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Lara Perryman
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Suzanne A Eccles
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Chris Jones
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, SM2 5NG, UK; Division of Molecular Pathology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Simon P Robinson
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, SM2 5NG, UK.
| |
Collapse
|
30
|
Anselmi M, Catalucci A, Felli V, Vellucci V, Di Sibio A, Gravina GL, Di Staso M, Di Cesare E, Masciocchi C. Diagnostic accuracy of proton magnetic resonance spectroscopy and perfusion-weighted imaging in brain gliomas follow-up: a single institutional experience. Neuroradiol J 2017. [PMID: 28627984 DOI: 10.1177/1971400916688354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Objectives The objective of this study was to evaluate whether proton magnetic resonance spectroscopy and perfusion magnetic resonance imaging (MRI) are able to increase diagnostic accuracy in the follow-up of brain gliomas, identifying the progression of disease before it becomes evident in the standard MRI; also to evaluate which of the two techniques has the best diagnostic accuracy. Methods Eighty-three patients with cerebral glioma (50 high-grade gliomas (HGGs), 33 low-grade gliomas (LGGs)) were retrospectively enrolled. All patients underwent standard MRI, H spectroscopic and perfusion echo-planar imaging MRI. For spectroscopy variations of choline/creatine, choline/N-acetyl-aspartate ratio, and lipids and lactates peak were considered. For perfusion 2.0 was considered the cerebral blood volume cut-off for progression. The combination of functional parameters gave a multiparametric score (0-2) to predict outcome. Diagnostic performance was determined by the receiver operating characteristic curve, with sensitivity, specificity, positive predictive and negative predictive values. Results In patients with LGGs a combined score of at least 1 was the best predictor for progression (odds ratio (OR) 3.91) with 8.4 months median anticipation of diagnosis compared to standard MRI. The individual advanced magnetic resonance technique did not show a diagnostic accuracy comparable to the combination of the two. Overall diagnostic accuracy area under the curve (AUC) was 0.881. In patients with HGGs the multiparametric score did not improve diagnostic accuracy significantly. Perfusion MRI was the best predictor of progression (OR 3.65), with 6.7 months median anticipation of diagnosis. Overall diagnostic accuracy AUC was 0.897. Then spectroscopy and perfusion MRI are able to identify tumour progression during follow-up earlier than standard MRI. Conclusion In patients with LGGs the combination of the functional parameters seems to be the best method for diagnosis of progression. In patients with HGGs perfusion is the best diagnostic method.
Collapse
Affiliation(s)
- Monica Anselmi
- 1 Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, San Salvatore Hospital of L'Aquila, Italy
| | - Alessia Catalucci
- 2 Division of Neuroradiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Valentina Felli
- 3 Division of Radiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Valentina Vellucci
- 3 Division of Radiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Alessandra Di Sibio
- 3 Division of Radiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Giovanni Luca Gravina
- 2 Division of Neuroradiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Mario Di Staso
- 4 Department of Radiotherapy, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Ernesto Di Cesare
- 3 Division of Radiology, Biotechnological and Applied Clinical Sciences, University of L'Aquila, Italy
| | - Carlo Masciocchi
- 1 Department of Biotechnology and Applied Clinical Sciences, University of L'Aquila, San Salvatore Hospital of L'Aquila, Italy
| |
Collapse
|
31
|
Seidu RA, Wu M, Su Z, Xu H. Paradoxical Role of High Mobility Group Box 1 in Glioma: A Suppressor or a Promoter? Oncol Rev 2017; 11:325. [PMID: 28382190 PMCID: PMC5364998 DOI: 10.4081/oncol.2017.325] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/13/2017] [Accepted: 02/27/2017] [Indexed: 01/08/2023] Open
Abstract
Gliomas represent 60% of primary intracranial brain tumors and 80% of all malignant types, with highest morbidity and mortality worldwide. Although glioma has been extensively studied, the molecular mechanisms underlying its pathology remain poorly understood. Clarification of the molecular mechanisms involved in their development and/or treatment resistance is highly required. High mobility group box 1 protein (HMGB1) is a nuclear protein that can also act as an extracellular trigger of inflammation, proliferation and migration, through receptor for advanced glycation end products and toll like receptors in a number of cancers including gliomas. It is known that excessive release of HMGB1 in cancer leads to unlimited replicative potential, ability to develop blood vessels (angiogenesis), evasion of programmed cell death (apoptosis), self-sufficiency in growth signals, insensitivity to inhibitors of growth, inflammation, tissue invasion and metastasis. In this review we explore the mechanisms by which HMGB1 regulates apoptosis and autophagy in glioma. We also looked at how HMGB1 mediates glioma regression and promotes angiogenesis as well as possible signaling pathways with an attempt to provide potential therapeutic targets for the treatment of glioma.
Collapse
Affiliation(s)
- Richard A. Seidu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University (Jiangbin Hospital), Zhenjiang, China
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Min Wu
- Department of Neurosurgery, Affiliated Hospital of Jiangsu University (Jiangbin Hospital), Zhenjiang, China
| | - Zhaoliang Su
- Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang, China
| |
Collapse
|
32
|
Damento GM, Koeller KK, Salomão DR, Pulido JS. T2 Fluid-Attenuated Inversion Recovery Imaging of Uveal Melanomas and Other Ocular Pathology. Ocul Oncol Pathol 2016; 2:251-261. [PMID: 27843906 DOI: 10.1159/000447265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/19/2016] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND/AIMS This study describes patterns of intraocular lesions on T2 fluid-attenuated inversion recovery (FLAIR) imaging, exploring a prospective role of FLAIR imaging sequence in diagnosis and treatment. METHODS A retrospective study of orbital magnetic resonance imaging (MRI) studies from the years 2000 to 2015 was performed. MRI sequences included: pre-contrast T1-weighted, T2-weighted, T2 FLAIR, and postcontrast T1 and T2 imaging gadolinium, which were evaluated by a neuroradiologist. Two cases of melanoma were correlated to their pathology. RESULTS Twenty-four patients with intraocular pathology were evaluated. All lesions, regardless of pigmentation, revealed previously described melanotic patterns on T1- and T2-weighted images; 80% of 10 melanomas localized were hyperintense on T2 FLAIR, which better delineated lesion margins. All of the four inflammatory pathologies on T2 FLAIR were hyperintense, as were 80% of the amelanotic neoplasms. Pathology of two large uveal melanomas paralleled the findings seen on T2 FLAIR. CONCLUSIONS T2 FLAIR appears beneficial in the demarcation of pigmented ocular lesions and may aid in determining protein content or previous treatment. Data also promote previous assertions that blood flow impacts intensity of lesions on T2 FLAIR. Further research is warranted.
Collapse
Affiliation(s)
- Gena M Damento
- Department of Ophthalmology, Mayo Clinic, Rochester, Minn., USA
| | | | - Diva R Salomão
- Department of Anatomic Pathology, Mayo Clinic, Rochester, Minn., USA
| | - Jose S Pulido
- Department of Ophthalmology, Mayo Clinic, Rochester, Minn., USA; Department of Molecular Medicine, Mayo Clinic, Rochester, Minn., USA
| |
Collapse
|
33
|
Bain AR, Nybo L, Ainslie PN. Cerebral Vascular Control and Metabolism in Heat Stress. Compr Physiol 2016; 5:1345-80. [PMID: 26140721 DOI: 10.1002/cphy.c140066] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
This review provides an in-depth update on the impact of heat stress on cerebrovascular functioning. The regulation of cerebral temperature, blood flow, and metabolism are discussed. We further provide an overview of vascular permeability, the neurocognitive changes, and the key clinical implications and pathologies known to confound cerebral functioning during hyperthermia. A reduction in cerebral blood flow (CBF), derived primarily from a respiratory-induced alkalosis, underscores the cerebrovascular changes to hyperthermia. Arterial pressures may also become compromised because of reduced peripheral resistance secondary to skin vasodilatation. Therefore, when hyperthermia is combined with conditions that increase cardiovascular strain, for example, orthostasis or dehydration, the inability to preserve cerebral perfusion pressure further reduces CBF. A reduced cerebral perfusion pressure is in turn the primary mechanism for impaired tolerance to orthostatic challenges. Any reduction in CBF attenuates the brain's convective heat loss, while the hyperthermic-induced increase in metabolic rate increases the cerebral heat gain. This paradoxical uncoupling of CBF to metabolism increases brain temperature, and potentiates a condition whereby cerebral oxygenation may be compromised. With levels of experimentally viable passive hyperthermia (up to 39.5-40.0 °C core temperature), the associated reduction in CBF (∼ 30%) and increase in cerebral metabolic demand (∼ 10%) is likely compensated by increases in cerebral oxygen extraction. However, severe increases in whole-body and brain temperature may increase blood-brain barrier permeability, potentially leading to cerebral vasogenic edema. The cerebrovascular challenges associated with hyperthermia are of paramount importance for populations with compromised thermoregulatory control--for example, spinal cord injury, elderly, and those with preexisting cardiovascular diseases.
Collapse
Affiliation(s)
- Anthony R Bain
- Centre for Heart Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Okanagan Campus, Kelowna, Canada
| | - Lars Nybo
- Department of Nutrition, Exercise and Sport Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Philip N Ainslie
- Centre for Heart Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia, Okanagan Campus, Kelowna, Canada
| |
Collapse
|
34
|
Zhang J, Liu C, Hou R. Knockdown of HMGB1 improves apoptosis and suppresses proliferation and invasion of glioma cells. Chin J Cancer Res 2014; 26:658-68. [PMID: 25561763 PMCID: PMC4279198 DOI: 10.3978/j.issn.1000-9604.2014.12.05] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 11/30/2014] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The purposes of this study were to explore the effects of high mobility group protein box 1 (HMGB1) gene on the growth, proliferation, apoptosis, invasion, and metastasis of glioma cells, with an attempt to provide potential therapeutic targets for the treatment of glioma. METHODS The expressions of HMGB1 in glioma cells (U251, U-87MG and LN-18) and one control cell line (SVG p12) were detected by real time PCR and Western blotting, respectively. Then, the effects of HMGB1 on the biological behaviors of glioma cells were detected: the expression of HMGB1 in human glioma cell lines U251 and U-87MG were suppressed using RNAi technique, then the influences of HMGB1 on the viability, cycle, apoptosis, and invasion abilities of U251 and U-87MG cells were analyzed using in a Transwell invasion chamber. Also, the effects of HMGB1 on the expressions of cyclin D1, Bax, Bcl-2, and MMP 9 were detected. RESULTS As shown by real-time PCR and Western blotting, the expression of HMGB1 significantly increased in glioma cells (U251, U-87MG, and LN-18) in comparison with the control cell line (SVG p12); the vitality, proliferation and invasive capabilities of U251 and U-87MG cells in the HMGB1 siRNA-transfected group were significantly lower than those in the blank control group and negative control (NC) siRNA group (P<0.05) but showed no significant difference between the blank control group and NC siRNA group. The percentage of apoptotic U251 and U-87MG cells was significantly higher in the HMGB1 siRNA-transfected group than in the blank control group and NC siRNA group (P<0.05) but was similar between the latter two groups. The HMGB1 siRNA-transfected group had significantly lower expression levels of Cyclin D1, Bcl-2, and MMP-9 protein in U251 and U-87MG cells and significantly higher expression of Bax protein than in the blank control group and NC siRNA group (P<0.05); the expression profiles of cyclin D1, Bax, Bcl-2, and MMP 9 showed no significant change in both blank control group and NC siRNA group. CONCLUSIONS HMGB1 gene may promote the proliferation and migration of glioma cells and suppress its effects of apoptosis. Inhibition of the expression of HMGB1 gene can suppress the proliferation and migration of glioma cells and promote their apoptosis. Our observations provided a new target for intervention and treatment of glioma.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Neurosurgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Cang Liu
- Department of Neurosurgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Ruiguang Hou
- Department of Neurosurgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| |
Collapse
|
35
|
Takagi H, Azuma K, Tsuka T, Imagawa T, Osaki T, Okamoto Y. Antitumor effects of high-temperature hyperthermia on a glioma rat model. Oncol Lett 2014; 7:1007-1010. [PMID: 24944659 PMCID: PMC3961387 DOI: 10.3892/ol.2014.1852] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 10/31/2013] [Indexed: 11/05/2022] Open
Abstract
High-temperature hyperthermia (HTH) is an established treatment option for cancer. The aim of the present study was to reveal the exact correlation between HTH at temperatures of 50-70°C and the resulting antitumor effects, using a glioma rat model. In the 60°C (T-60) and 70°C (T-70) HTH groups, tumor growth rates were significantly suppressed compared with those in the nontreatment (NT) group. In the 50°C (T-50) HTH group, tumor growth rates were not suppressed compared with those in the NT group. The numbers of terminal dUTP nick-end labeling-positive cells in tumor tissue were significantly higher in the T-50, T-60 and T-70 groups than those in the NT group. The Ki-67-positive areas were significantly decreased in the T-70 group compared with those in the NT and T-60 groups. Our data indicate that HTH at 60 and 70°C suppresses tumor growth in a glioma rat model. In particular, cell proliferation was significantly suppressed by HTH at 70°C. However, differences in the mechanism of action of HTH at 60 and 70°C were observed.
Collapse
Affiliation(s)
- Hidefumi Takagi
- The United Graduate School of Veterinary Science, Yamaguchi University, Yamaguchi 753-8515, Japan ; Takagi Animal Clinic, Saijo, Ehime 793-0035, Japan
| | - Kazuo Azuma
- Faculty of Agriculture, Tottori University, Tottori 680-8533, Japan
| | - Takeshi Tsuka
- Faculty of Agriculture, Tottori University, Tottori 680-8533, Japan
| | - Tomohiro Imagawa
- Faculty of Agriculture, Tottori University, Tottori 680-8533, Japan
| | - Tomohiro Osaki
- Faculty of Agriculture, Tottori University, Tottori 680-8533, Japan
| | | |
Collapse
|
36
|
Rossmeisl JH, Garcia PA, Daniel GB, Bourland JD, Debinski W, Dervisis N, Klahn S. Invited review--neuroimaging response assessment criteria for brain tumors in veterinary patients. Vet Radiol Ultrasound 2013; 55:115-32. [PMID: 24219161 DOI: 10.1111/vru.12118] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 09/07/2013] [Indexed: 12/28/2022] Open
Abstract
The evaluation of therapeutic response using cross-sectional imaging techniques, particularly gadolinium-enhanced MRI, is an integral part of the clinical management of brain tumors in veterinary patients. Spontaneous canine brain tumors are increasingly recognized and utilized as a translational model for the study of human brain tumors. However, no standardized neuroimaging response assessment criteria have been formulated for use in veterinary clinical trials. Previous studies have found that the pathophysiologic features inherent to brain tumors and the surrounding brain complicate the use of the response evaluation criteria in solid tumors (RECIST) assessment system. Objectives of this review are to describe strengths and limitations of published imaging-based brain tumor response criteria and propose a system for use in veterinary patients. The widely used human Macdonald and response assessment in neuro-oncology (RANO) criteria are reviewed and described as to how they can be applied to veterinary brain tumors. Discussion points will include current challenges associated with the interpretation of brain tumor therapeutic responses such as imaging pseudophenomena and treatment-induced necrosis, and how advancements in perfusion imaging, positron emission tomography, and magnetic resonance spectroscopy have shown promise in differentiating tumor progression from therapy-induced changes. Finally, although objective endpoints such as MR imaging and survival estimates will likely continue to comprise the foundations for outcome measures in veterinary brain tumor clinical trials, we propose that in order to provide a more relevant therapeutic response metric for veterinary patients, composite response systems should be formulated and validated that combine imaging and clinical assessment criteria.
Collapse
Affiliation(s)
- John H Rossmeisl
- Department of Small Animal Clinical Sciences, Virginia-Maryland Regional College of Veterinary Medicine, VA, 24061; Biomechanical Systems and Veterinary and Comparative Neuro-oncology Laboratories, Department of Biomedical Engineering, Virginia Tech-Wake Forest University School of Biomedical Engineering and Sciences, VA, 24061
| | | | | | | | | | | | | |
Collapse
|
37
|
Jackson A. The changing face of brain tumours. Br J Radiol 2011; 84 Spec No 2:S79-81. [DOI: 10.1259/bjr/92477419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|