1
|
Small SL. Precision neurology. Ageing Res Rev 2025; 104:102632. [PMID: 39657848 DOI: 10.1016/j.arr.2024.102632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/23/2024] [Accepted: 12/05/2024] [Indexed: 12/12/2024]
Abstract
Over the past several decades, high-resolution brain imaging, blood and cerebrospinal fluid analyses, and other advanced technologies have changed diagnosis from an exercise depending primarily on the history and physical examination to a computer- and online resource-aided process that relies on larger and larger quantities of data. In addition, randomized controlled trials (RCT) at a population level have led to many new drugs and devices to treat neurological disease, including disease-modifying therapies. We are now at a crossroads. Combinatorially profound increases in data about individuals has led to an alternative to population-based RCTs. Genotyping and comprehensive "deep" phenotyping can sort individuals into smaller groups, enabling precise medical decisions at a personal level. In neurology, precision medicine that includes prediction, prevention and personalization requires that genomic and phenomic information further incorporate imaging and behavioral data. In this article, we review the genomic, phenomic, and computational aspects of precision medicine for neurology. After defining biological markers, we discuss some applications of these "-omic" and neuroimaging measures, and then outline the role of computation and ultimately brain simulation. We conclude the article with a discussion of the relation between precision medicine and value-based care.
Collapse
Affiliation(s)
- Steven L Small
- Department of Neuroscience, University of Texas at Dallas, Dallas, TX, USA; Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurology, The University of Chicago, Chicago, IL, USA; Department of Neurology, University of California, Irvine, Orange, CA, USA.
| |
Collapse
|
2
|
Pérez-Alfayate R, Cabezas-Camarero S, Castaño-Montoya JP, Arévalo-Saenz AA, Carrascosa-Granada A, Alonso-Lera P, Grasso G. Communicating hydrocephalus in glioblastoma presenting as chronic hydrocephalus: systematic review and meta-analysis. Acta Neurochir (Wien) 2025; 167:19. [PMID: 39820650 PMCID: PMC11739179 DOI: 10.1007/s00701-024-06414-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE Communicating hydrocephalus (CH) is an uncommon complication that can affect patients with glioblastoma (GBM). Due to its clinical and radiological findings, it presents as a chronic hydrocephalus. The mechanisms underlying its occurrence and impact on the prognosis of the disease are poorly known, but some studies have suggested that shunting can have a positive impact on the quality of life of these patients. METHODS The authors performed a systematic literature review and meta-analysis to identify the possible risk factors that could help to identify CH cases in glioblastoma, using the MEDLINE/PubMed and Cochrane Database of Systematic Reviews databases. The Joanna Briggs Institute critical appraisal tool was used to assess the risk of bias. RESULTS Our search yielded 273 studies, but only 9 records were included in the final quantitative analysis. CH in glioblastoma patients was found to be very uncommon (proportion 0.04 out of 1; range 0.03-0.05, p < 0.01, CI:95%) and its occurrence was associated with a previous ventricular opening (0.85 out of 1; range 0.66-0.94, p < 0.01, CI:95%). CONCLUSION CH in glioblastoma should be suspected in patients with GBM who have undergone accidental ventricular opening during tumor resection and presenting with chronic hydrocephalus symptoms.
Collapse
Affiliation(s)
| | | | | | | | | | - Pedro Alonso-Lera
- Neurosurgical Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Giovanni Grasso
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), Neurosurgical Unit, University of Palermo, Palermo, Italy
| |
Collapse
|
3
|
Nguyen DH, Nguyen DH, Le TD, Nguyen HK, Nguyen-Thi VA, Nguyen MD. Diagnostic algorithm for glioma grading using dynamic susceptibility contrast‑enhanced magnetic resonance perfusion and proton magnetic resonance spectroscopy. Biomed Rep 2024; 20:56. [PMID: 38357240 PMCID: PMC10865167 DOI: 10.3892/br.2024.1741] [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: 07/17/2023] [Accepted: 12/14/2023] [Indexed: 02/16/2024] Open
Abstract
The present retrospective study aimed to investigate the diagnostic capacity of and design a diagnostic algorithm for dynamic susceptibility contrast-enhanced MRI (DSCE-MRI) and proton magnetic resonance spectroscopy (1H-MRS) in grading low-grade glioma (LGG) and high-grade glioma (HGG). This retrospective study enrolled 57 patients, of which 14 had LGG and 43 had HGG, five had World Health Organization grade 1, nine had grade 2, 20 had grade 3 and 23 had grade 4 glioma. All subjects underwent a standard 3T MRI brain tumor protocol with conventional MRI (cMRI) and advanced techniques, including DSCE-MRI and 1H-MRS. The associations of grade categorization with parameters in tumor and peritumor regions in the DSCE-MRI were examined, including tumor relative cerebral blood volume (TrCBV) and peripheral relative (Pr)CBV, as well as Tr and Pr cerebral blood flow (CBF) and 1H-MRS, including the creatine (Cr) and N-acetyl aspartate (NAA) ratios of choline (Cho), i.e. the TCho/NAA, PCho/NAA, TCho/Cr and PCho/Cr metabolite ratios. The data were compared using the Mann-Whitney U-test, independent samples t-test, Chi-square test, Fisher's exact test and receiver operating characteristic curve analyses. Decision tree analysis established an algorithm based on cutoffs for specified significant parameters. The PrCBF had the highest performance in the preoperative prediction of histological glioma grading, followed by the TrCBV, PrCBF, TrCBV, PCho/NAA, PCho/Cr, TCho/NAA and TCho/Cr. An algorithm based on TrCBV, PrCBF and TCho/Cr had a diagnostic accuracy of 100% for LGG and 90.7% for HGG and a misclassification risk of 7%. The cutoffs (sensitivity and specificity) were 2.48 (86 and 100%) for TrCBV, 1.26 (83.7 and 100%) for PrCBF and 3.18 (69.8 and 78.6%) for TCho/Cr. In conclusion, the diagnostic algorithm using TrCBV, PrCBF and TCho/Cr values, which were obtained from DSCE-MRI and 1H-MRS, increased diagnostic accuracy to 100% for LGGs and 90.7% for HGGs compared to previous studies using conventional MRI. This non-invasive advanced MRI diagnostic algorithm is recommended for clinical application for constructing preoperative strategies and prognosis of patients with glioma.
Collapse
Affiliation(s)
- Dinh Hieu Nguyen
- Department of Radiology, Hanoi Medical University, Hanoi 100000, Vietnam
- Department of Radiology, Ha Dong General Hospital, Hanoi 100000, Vietnam
| | - Duy Hung Nguyen
- Department of Radiology, Hanoi Medical University, Hanoi 100000, Vietnam
- Department of Radiology, Viet Duc Hospital, Hanoi 100000, Vietnam
| | - Thanh Dung Le
- Department of Radiology, Viet Duc Hospital, Hanoi 100000, Vietnam
- Department of Radiology, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi 100000, Vietnam
| | - Ha Khuong Nguyen
- Department of Radiology, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Van Anh Nguyen-Thi
- Department of Radiology, Hanoi Medical University Hospital, Hanoi 100000, Vietnam
| | - Minh Duc Nguyen
- Department of Radiology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City 700000, Vietnam
| |
Collapse
|
4
|
Dung LT, Hung ND, Hung NK, Khuong NH, Thien LQ, Duy NQ, Duc NM. Magnetic resonance imaging characteristics of glioblastoma of the optic pathway during adulthood. Radiol Case Rep 2023; 18:2628-2632. [PMID: 37273722 PMCID: PMC10238590 DOI: 10.1016/j.radcr.2023.05.010] [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: 03/01/2023] [Revised: 04/29/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Glioma has been previously known as the most common adult brain tumor. Glioma of the optic pathway is predominated by low-grade neoplasms. High-grade glioma in adults is extremely rare. In this study, we present the case of a 46-year-old male patient who developed glioblastoma of the optic chiasm extending along the optic tract. This study aims to discuss several common differential diagnoses of nontumor diffuse lesions in the optic pathway and their clinical symptoms and magnetic resonance imaging findings, which could help navigate management.
Collapse
Affiliation(s)
- Le Thanh Dung
- Department of Radiology, VNU University of Medicine and Pharmacy, Vietnam National University, Hanoi, Viet Nam
- Department of Radiology, Viet Duc Hospital, Hanoi, Viet Nam
| | - Nguyen Duy Hung
- Department of Radiology, Viet Duc Hospital, Hanoi, Viet Nam
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Nguyen Khac Hung
- Department of Radiology, Hanoi Obstetrics and Gynecology, Hanoi, Viet Nam
| | - Nguyen Ha Khuong
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Le Quy Thien
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
| | - Ngo Quang Duy
- Department of Radiology, Hanoi Medical University, Hanoi, Viet Nam
- Department of Radiology, Ha Giang General Hospital, Ha Giang, Viet Nam
| | - Nguyen Minh Duc
- Department of Radiology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Viet Nam
| |
Collapse
|
5
|
Calabrese E, Rudie JD, Rauschecker AM, Villanueva-Meyer JE, Clarke JL, Solomon DA, Cha S. Combining radiomics and deep convolutional neural network features from preoperative MRI for predicting clinically relevant genetic biomarkers in glioblastoma. Neurooncol Adv 2022; 4:vdac060. [PMID: 35611269 PMCID: PMC9122791 DOI: 10.1093/noajnl/vdac060] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Glioblastoma is the most common primary brain malignancy, yet treatment options are limited, and prognosis remains guarded. Individualized tumor genetic assessment has become important for accurate prognosis and for guiding emerging targeted therapies. However, challenges remain for widespread tumor genetic testing due to costs and the need for tissue sampling. The aim of this study is to evaluate a novel artificial intelligence method for predicting clinically relevant genetic biomarkers from preoperative brain MRI in patients with glioblastoma.
Methods
We retrospectively analyzed preoperative MRI data from 400 patients with glioblastoma and grade 4 astrocytoma, IDH mutant who underwent resection and genetic testing. Nine genetic biomarkers were assessed: hotspot mutations of IDH1 or TERT promoter, pathogenic mutations of TP53, PTEN, ATRX, or CDKN2A/B, MGMT promoter methylation, EGFR amplification, and combined aneuploidy of chromosomes 7 & 10. Models were developed to predict biomarker status from MRI data using radiomics features, convolutional neural network (CNN) features, and a combination of both.
Results
Combined model performance was good for IDH1 and TERT promoter hotspot mutations, pathogenic mutations of ATRX and CDKN2A/B, and combined aneuploidy of chromosomes 7 & 10, with receiver operating characteristic area under the curve (ROC AUC) > 0.85 and was fair for all other tested biomarkers with ROC AUC > 0.7. Combined model performance was statistically superior to individual radiomics and CNN feature models for prediction chromosome 7 & 10 aneuploidy, MGMT promoter methylation, and PTEN loss.
Conclusions
Combining radiomics and CNN features from preoperative MRI yields improved non-invasive genetic biomarker prediction performance in patients with grade 4 diffuse gliomas.
Collapse
Affiliation(s)
- Evan Calabrese
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
- Center for Intelligent Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey D Rudie
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| | - Andreas M Rauschecker
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
- Center for Intelligent Imaging, University of California San Francisco, San Francisco, California, USA
| | - Jennifer L Clarke
- Division of Neuro-Oncology, Department of Neurology and Neurological Surgery, University of California San Francisco, San Francisco, California, USA
| | - David A Solomon
- Department of Pathology, University of California San Francisco, San Francisco, California, USA
- Clinical Cancer Genomics Laboratory, University of California San Francisco, San Francisco, California, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, California, USA
| |
Collapse
|
6
|
El Rahal A, Cipriani D, Fung C, Hohenhaus M, Sveikata L, Straehle J, Shah MJ, Heiland HD, Beck J, Schnell O. Hydrocephalus Shunting in Supratentorial Glioblastoma: Functional Outcomes and Management. Front Oncol 2022; 12:796105. [PMID: 35223477 PMCID: PMC8865077 DOI: 10.3389/fonc.2022.796105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Background Glioblastoma is the most common and the most challenging to treat adult primary central nervous system tumor. Although modern management strategies modestly improved the overall survival, the prognosis remains dismal associated with poor life quality and the clinical course often dotted by treatment side effects and cognitive decline. Functional deterioration might be caused by obstructive or communicating hydrocephalus but due to poor overall prognosis surgical treatment options are often limited and its optimal management strategies remain elusive. We aimed to investigate risk factors, treatment options and outcomes for tumor-associated hydrocephalus in a contemporary 10 years cohort of glioblastoma patients. Methods We reviewed electronic health records of 1800 glioblastoma patients operated at the Department of Neurosurgery, Medical Center – University of Freiburg from 2009 to 2019. Demographics, clinical characteristics and radiological features were analyzed. Univariate analysis for nominal variables was performed either by Fisher’s exact test or Chi-square test, as appropriate. Results We identified 39 glioblastoma patients with symptomatic communicating hydrocephalus treated by ventricular shunting (incidence 2.1%). Opening of the ventricular system during a previous tumor resection was associated with symptomatic hydrocephalus (p<0.05). There was also a trend toward location (frontal and temporal) and larger tumor volume. Number of craniotomies before shunting was not considered as a risk factor. Shunting improved hydrocephalus symptoms in 95% of the patients and Karnofsky Performance Score (KPS) could be restored after shunting. Of note, 75% of the patients had a post-shunting oncological treatment such as radiotherapy or chemotherapy, most prevalently chemotherapy. Infection (7.7%) and over- or under drainage (17.9%) were the most common complications requiring shunt revision in ten patients (25.6%), No peritoneal metastasis was found. The median overall survival (OS) was 385 days and the median post shunting survival was 135 days. Conclusion Ventricular system opening was identified as a risk factor for communicating hydrocephalus in glioblastoma patients. Although glioblastoma treatment remains challenging, shunting improved hydrocephalus-related functional status and may be considered even in a palliative setting for symptom relief.
Collapse
Affiliation(s)
- Amir El Rahal
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany.,Department of Neurosurgery, Department of Clinical Neurosciences, Geneva University Hospitals, Faculty of Medicine, Geneva, Switzerland
| | - Debora Cipriani
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Christian Fung
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Marc Hohenhaus
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Lukas Sveikata
- J.P. Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States.,Division of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, Geneva, Switzerland
| | - Jakob Straehle
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Mukesch Johannes Shah
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Henrik Dieter Heiland
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Jürgen Beck
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center - University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
7
|
Defining the impact of adjuvant treatment on the prognosis of patients with inoperable glioblastoma undergoing biopsy only: does the survival benefit outweigh the treatment effort? Neurosurg Rev 2022; 45:2339-2347. [PMID: 35194724 PMCID: PMC9160139 DOI: 10.1007/s10143-022-01754-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/02/2022]
Abstract
Patients with inoperable glioblastoma (GBM) usually experience worse prognosis compared to those in whom gross total resection (GTR) is achievable. Considering the treatment duration and its side effects identification of patients with survival benefit from treatment is essential to guarantee the best achievable quality of life. The aim of this study was to evaluate the survival benefit from radio-chemotherapy and to identify clinical, molecular, and imaging parameters associated with better outcome in patients with biopsied GBMs. Consecutive patients with inoperable GBM who underwent tumor biopsy at our department from 2005 to 2019 were retrospectively analyzed. All patients had histologically confirmed GBM and were followed up until death. The overall survival (OS) was calculated from date of diagnosis to date of death. Clinical, radiological, and molecular predictors of OS were evaluated. A total of 95 patients with biopsied primary GBM were enrolled in the study. The mean age was 64.3 ± 13.2 years; 56.8% (54/95) were male, and 43.2% (41/95) female. Median OS in the entire cohort was 5.5 months. After stratification for adjuvant treatment, a higher median OS was found in the group with adjuvant treatment (7 months, range 2–88) compared to the group without treatment (1 month, range 1–5) log-rank test, p < 0.0001. Patients with inoperable GBM undergoing biopsy indeed experience a very limited OS. Adjuvant treatment is associated with significantly longer OS compared to patients not receiving treatment and should be considered, especially in younger patients with good clinical condition at presentation.
Collapse
|
8
|
Vessières A, Quissac E, Lemaire N, Alentorn A, Domeracka P, Pigeon P, Sanson M, Idbaih A, Verreault M. Heterogeneity of Response to Iron-Based Metallodrugs in Glioblastoma Is Associated with Differences in Chemical Structures and Driven by FAS Expression Dynamics and Transcriptomic Subtypes. Int J Mol Sci 2021; 22:ijms221910404. [PMID: 34638742 PMCID: PMC8508975 DOI: 10.3390/ijms221910404] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 11/18/2022] Open
Abstract
Glioblastoma (GBM) is the most frequent and deadliest primary brain cancer in adults, justifying the search for new treatments. Some members of the iron-based ferrocifen family have demonstrated a high cytotoxic effect on various cancer cell lines via innovative mechanisms of action. Here, we evaluated the antiproliferative activity by wst-1 assay of six ferrocifens in 15 molecularly diverse GBM patient-derived cell lines (PDCLs). In five out of six compounds, the half maximal inhibitory concentration (IC50) values varied significantly (10 nM < IC50 < 29.8 µM) while the remaining one (the tamoxifen-like complex) was highly cytotoxic against all PDCLs (mean IC50 = 1.28 µM). The pattern of response was comparable for the four ferrocifens bearing at least one phenol group and differed widely from those of the tamoxifen-like complex and the complex with no phenol group. An RNA sequencing differential analysis showed that response to the diphenol ferrocifen relied on the activation of the Death Receptor signaling pathway and the modulation of FAS expression. Response to this complex was greater in PDCLs from the Mesenchymal or Proneural transcriptomic subtypes compared to the ones from the Classical subtype. These results provide new information on the mechanisms of action of ferrocifens and highlight a broader diversity of behavior than previously suspected among members of this family. They also support the case for a molecular-based personalized approach to future use of ferrocifens in the treatment of GBM.
Collapse
Affiliation(s)
- Anne Vessières
- Institut Parisien de Chimie Moléculaire, Sorbonne Université, CNRS, UMR CNRS 8232, 4 Place Jussieu, F-75005 Paris, France;
- Correspondence: (A.V.); (M.V.)
| | - Emie Quissac
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France; (E.Q.); (N.L.); (P.D.)
| | - Nolwenn Lemaire
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France; (E.Q.); (N.L.); (P.D.)
| | - Agusti Alentorn
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Service de Neurologie 2-Mazarin, F-75013 Paris, France; (A.A.); (M.S.); (A.I.)
| | - Patrycja Domeracka
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France; (E.Q.); (N.L.); (P.D.)
| | - Pascal Pigeon
- Institut Parisien de Chimie Moléculaire, Sorbonne Université, CNRS, UMR CNRS 8232, 4 Place Jussieu, F-75005 Paris, France;
- Chimie ParisTech-PSL, 11 Rue P. et M. Curie, F-75005 Paris, France
| | - Marc Sanson
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Service de Neurologie 2-Mazarin, F-75013 Paris, France; (A.A.); (M.S.); (A.I.)
| | - Ahmed Idbaih
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, DMU Neurosciences, Service de Neurologie 2-Mazarin, F-75013 Paris, France; (A.A.); (M.S.); (A.I.)
| | - Maïté Verreault
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm, Sorbonne Université, CNRS, APHP, Hôpital de la Pitié Salpêtrière, F-75013 Paris, France; (E.Q.); (N.L.); (P.D.)
- Correspondence: (A.V.); (M.V.)
| |
Collapse
|
9
|
Ashrafizadeh M, Zarabi A, Hushmandi K, Moghadam ER, Hashemi F, Daneshi S, Hashemi F, Tavakol S, Mohammadinejad R, Najafi M, Dudha N, Garg M. C-Myc Signaling Pathway in Treatment and Prevention of Brain Tumors. Curr Cancer Drug Targets 2021; 21:2-20. [PMID: 33069197 DOI: 10.2174/1568009620666201016121005] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/26/2020] [Accepted: 09/07/2020] [Indexed: 12/16/2022]
Abstract
Brain tumors are responsible for high morbidity and mortality worldwide. Several factors such as the presence of blood-brain barrier (BBB), sensitive location in the brain, and unique biological features challenge the treatment of brain tumors. The conventional drugs are no longer effective in the treatment of brain tumors, and scientists are trying to find novel therapeutics for brain tumors. In this way, identification of molecular pathways can facilitate finding an effective treatment. c-Myc is an oncogene signaling pathway capable of regulation of biological processes such as apoptotic cell death, proliferation, survival, differentiation, and so on. These pleiotropic effects of c-Myc have resulted in much fascination with its role in different cancers, particularly brain tumors. In the present review, we aim to demonstrate the upstream and down-stream mediators of c-Myc in brain tumors such as glioma, glioblastoma, astrocytoma, and medulloblastoma. The capacity of c-Myc as a prognostic factor in brain tumors will be investigated. Our goal is to define an axis in which the c-Myc signaling pathway plays a crucial role and to provide direction for therapeutic targeting in these signaling networks in brain tumors.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Universite Caddesi No. 27, Orhanli, Tuzla, 34956 Istanbul, Turkey
| | - Ali Zarabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farid Hashemi
- DVM. Graduated, Young Researcher and Elite Club, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of physiotherapy, Faculty of rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Reza Mohammadinejad
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7619813159, Iran
| | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Namrata Dudha
- Department of Biotechnology and Microbiology, School of Sciences, Noida International University, Gautam Budh Nagar, Uttar Pradesh, India
| | - Manoj Garg
- Amity of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida-201313, India
| |
Collapse
|
10
|
Li D, Patel CB, Xu G, Iagaru A, Zhu Z, Zhang L, Cheng Z. Visualization of Diagnostic and Therapeutic Targets in Glioma With Molecular Imaging. Front Immunol 2020; 11:592389. [PMID: 33193439 PMCID: PMC7662122 DOI: 10.3389/fimmu.2020.592389] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/08/2020] [Indexed: 02/04/2023] Open
Abstract
Gliomas, particularly high-grade gliomas including glioblastoma (GBM), represent the most common and malignant types of primary brain cancer in adults, and carry a poor prognosis. GBM has been classified into distinct subgroups over the years based on cellular morphology, clinical characteristics, biomarkers, and neuroimaging findings. Based on these classifications, differences in therapeutic response and patient outcomes have been established. Recently, the identification of complex molecular signatures of GBM has led to the development of diverse targeted therapeutic regimens and translation into multiple clinical trials. Chemical-, peptide-, antibody-, and nanoparticle-based probes have been designed to target specific molecules in gliomas and then be visualized with multimodality molecular imaging (MI) techniques including positron emission tomography (PET), single-photon emission computed tomography (SPECT), near-infrared fluorescence (NIRF), bioluminescence imaging (BLI), and magnetic resonance imaging (MRI). Thus, multiple molecules of interest can now be noninvasively imaged to guide targeted therapies with a potential survival benefit. Here, we review developments in molecular-targeted diagnosis and therapy in glioma, MI of these targets, and MI monitoring of treatment response, with a focus on the biological mechanisms of these advanced molecular probes. MI probes have the potential to noninvasively demonstrate the pathophysiologic features of glioma for diagnostic, treatment, and response assessment considerations for various targeted therapies, including immunotherapy. However, most MI tracers are in preclinical development, with only integrin αVβ3 and isocitrate dehydrogenase (IDH)-mutant MI tracers having been translated to patients. Expanded international collaborations would accelerate translational research in the field of glioma MI.
Collapse
Affiliation(s)
- Deling Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
| | - Chirag B. Patel
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
- Division of Neuro-Oncology, Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA, United States
| | - Guofan Xu
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Andrei Iagaru
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Zhaohui Zhu
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Department of Radiology, School of Medicine, Stanford University, Stanford, CA, United States
| |
Collapse
|
11
|
Calabrese E, Villanueva-Meyer JE, Cha S. A fully automated artificial intelligence method for non-invasive, imaging-based identification of genetic alterations in glioblastomas. Sci Rep 2020; 10:11852. [PMID: 32678261 PMCID: PMC7366666 DOI: 10.1038/s41598-020-68857-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/29/2020] [Indexed: 02/02/2023] Open
Abstract
Glioblastoma is the most common malignant brain parenchymal tumor yet remains challenging to treat. The current standard of care-resection and chemoradiation-is limited in part due to the genetic heterogeneity of glioblastoma. Previous studies have identified several tumor genetic biomarkers that are frequently present in glioblastoma and can alter clinical management. Currently, genetic biomarker status is confirmed with tissue sampling, which is costly and only available after tumor resection or biopsy. The purpose of this study was to evaluate a fully automated artificial intelligence approach for predicting the status of several common glioblastoma genetic biomarkers on preoperative MRI. We retrospectively analyzed multisequence preoperative brain MRI from 199 adult patients with glioblastoma who subsequently underwent tumor resection and genetic testing. Radiomics features extracted from fully automated deep learning-based tumor segmentations were used to predict nine common glioblastoma genetic biomarkers with random forest regression. The proposed fully automated method was useful for predicting IDH mutations (sensitivity = 0.93, specificity = 0.88), ATRX mutations (sensitivity = 0.94, specificity = 0.92), chromosome 7/10 aneuploidies (sensitivity = 0.90, specificity = 0.88), and CDKN2 family mutations (sensitivity = 0.76, specificity = 0.86).
Collapse
Affiliation(s)
- Evan Calabrese
- Department of Radiology and Biomedical Imaging, University of California At San Francisco, 350 Parnassus Ave, Suite 307H, San Francisco, CA, 94143-0628, USA.
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California At San Francisco, 350 Parnassus Ave, Suite 307H, San Francisco, CA, 94143-0628, USA
| | - Soonmee Cha
- Department of Radiology and Biomedical Imaging, University of California At San Francisco, 350 Parnassus Ave, Suite 307H, San Francisco, CA, 94143-0628, USA
| |
Collapse
|
12
|
Ene CI, Macomber MW, Barber JK, Ferreira MJ, Ellenbogen RG, Holland EC, Rockhill JK, Silbergeld DL, Halasz LM. Patterns of Failure After Stereotactic Radiosurgery for Recurrent High-Grade Glioma: A Single Institution Experience of 10 Years. Neurosurgery 2020; 85:E322-E331. [PMID: 30576476 DOI: 10.1093/neuros/nyy520] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 10/04/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Stereotactic radiosurgery (SRS) is a treatment modality that is frequently used as salvage therapy for small nodular recurrent high-grade gliomas (HGG). Due to the infiltrative nature of HGG, it is unclear if this highly focused technique provides a durable local control benefit. OBJECTIVE To determine how demographic or clinical factors influence the pattern of failure following SRS for recurrent high-grade gliomas. METHODS We retrospectively reviewed clinical, radiographic, and follow-up information for 47 consecutive patients receiving SRS for recurrent HGG at our institution between June 2006 and July 2016. All patients initially presented with an HGG (WHO grade III and IV). Following SRS for recurrence, all patients experienced treatment failure, and we evaluated patterns of local, regional, and distant failure in relation to the SRS 50% isodose line. RESULTS Most patients with recurrent HGG developed "in-field" treatment failure following SRS (n = 40; 85%). Higher SRS doses were associated with longer time to failure (hazards ratio = 0.80 per 1 Gy increase; 95% confidence interval 0.67-0.96; P = .016). There was a statistically significant increase in distant versus in-field failure among older patients (P = .035). This effect was independent of bevacizumab use (odds ratio = 0.54, P = 1.0). CONCLUSION Based on our experience, the majority of treatment failures after SRS for recurrent HGG were "in-field." Older patients, however, presented with more distant failures. Our results indicate that higher SRS doses delivered to a larger area as fractioned or unfractioned regimen may prolong time to failure, especially in the older population.
Collapse
Affiliation(s)
- Chibawanye I Ene
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Meghan W Macomber
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Jason K Barber
- Department of Neurological Surgery, University of Washington, Seattle, Washington
| | - Manuel J Ferreira
- Department of Neurological Surgery, University of Washington, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Richard G Ellenbogen
- Department of Neurological Surgery, University of Washington, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Eric C Holland
- Department of Neurological Surgery, University of Washington, Seattle, Washington.,Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Jason K Rockhill
- Department of Radiation Oncology, University of Washington, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Daniel L Silbergeld
- Department of Neurological Surgery, University of Washington, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| | - Lia M Halasz
- Department of Radiation Oncology, University of Washington, Seattle, Washington.,Alvord Brain Tumor Center, University of Washington, Seattle, Washington
| |
Collapse
|
13
|
Juthani R, Madajewski B, Yoo B, Zhang L, Chen PM, Chen F, Turker MZ, Ma K, Overholtzer M, Longo VA, Carlin S, Aragon-Sanabria V, Huse J, Gonen M, Zanzonico P, Rudin CM, Wiesner U, Bradbury MS, Brennan CW. Ultrasmall Core-Shell Silica Nanoparticles for Precision Drug Delivery in a High-Grade Malignant Brain Tumor Model. Clin Cancer Res 2019; 26:147-158. [PMID: 31515460 DOI: 10.1158/1078-0432.ccr-19-1834] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/25/2019] [Accepted: 09/09/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Small-molecule inhibitors have revolutionized treatment of certain genomically defined solid cancers. Despite breakthroughs in treating systemic disease, central nervous system (CNS) metastatic progression is common, and advancements in treating CNS malignancies remain sparse. By improving drug penetration across a variably permeable blood-brain barrier and diffusion across intratumoral compartments, more uniform delivery and distribution can be achieved to enhance efficacy. EXPERIMENTAL DESIGN Ultrasmall fluorescent core-shell silica nanoparticles, Cornell prime dots (C' dots), were functionalized with αv integrin-binding (cRGD), or nontargeting (cRAD) peptides, and PET labels (124I, 89Zr) to investigate the utility of dual-modality cRGD-C' dots for enhancing accumulation, distribution, and retention (ADR) in a genetically engineered mouse model of glioblastoma (mGBM). mGBMs were systemically treated with 124I-cRGD- or 124I-cRAD-C' dots and sacrificed at 3 and 96 hours, with concurrent intravital injections of FITC-dextran for mapping blood-brain barrier breakdown and the nuclear stain Hoechst. We further assessed target inhibition and ADR following attachment of dasatinib, creating nanoparticle-drug conjugates (Das-NDCs). Imaging findings were confirmed with ex vivo autoradiography, fluorescence microscopy, and p-S6RP IHC. RESULTS Improvements in brain tumor delivery and penetration, as well as enhancement in the ADR, were observed following administration of integrin-targeted C' dots, as compared with a nontargeted control. Furthermore, attachment of the small-molecule inhibitor, dasatinib, led to its successful drug delivery throughout mGBM, demonstrated by downstream pathway inhibition. CONCLUSIONS These results demonstrate that highly engineered C' dots are promising drug delivery vehicles capable of navigating the complex physiologic barriers observed in a clinically relevant brain tumor model.
Collapse
Affiliation(s)
- Rupa Juthani
- Department of Neurosurgery, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Brian Madajewski
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Barney Yoo
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York. .,Department of Chemistry, Hunter College, The City University of New York, New York, New York
| | - Li Zhang
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Pei-Ming Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Feng Chen
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Melik Z Turker
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York
| | - Kai Ma
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York
| | - Michael Overholtzer
- Cell Biology Program, Sloan Kettering Institute for Cancer Research, New York, New York.,BCMB Allied Program, Weill Cornell Medical College, New York, New York
| | - Valerie A Longo
- Small-Animal Imaging Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean Carlin
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York
| | | | - Jason Huse
- Human Oncology & Pathogenesis Program, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Pat Zanzonico
- Department of Medical Physics, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Charles M Rudin
- Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ulrich Wiesner
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York.
| | - Michelle S Bradbury
- Department of Radiology, Sloan Kettering Institute for Cancer Research, New York, New York. .,Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, New York, New York
| | - Cameron W Brennan
- Department of Neurosurgery, Sloan Kettering Institute for Cancer Research, New York, New York.
| |
Collapse
|
14
|
Petrova L, Korfiatis P, Petr O, LaChance DH, Parney I, Buckner JC, Erickson BJ. Cerebral blood volume and apparent diffusion coefficient - Valuable predictors of non-response to bevacizumab treatment in patients with recurrent glioblastoma. J Neurol Sci 2019; 405:116433. [PMID: 31476621 DOI: 10.1016/j.jns.2019.116433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/30/2019] [Accepted: 08/22/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. The core of standard of care for newly diagnosed GBM was established in 2005 and includes maximum feasible surgical resection followed by radiation and temozolomide, with subsequent temozolomide with or without tumor-treating fields. Unfortunately, nearly all patients experience a recurrence. Bevacizumab (BV) is a commonly used second-line agent for such recurrences, but it has not been shown to impact overall survival, and short-term response is variable. METHODS We collected MRI perfusion and diffusion images from 54 subjects with recurrent GBM treated only with radiation and temozolomide. They were subsequently treated with BV. Using machine learning, we created a model to predict short term response (6 months) and overall survival. We set time thresholds to maximize the separation of responders/survivors versus non-responders/short survivors. RESULTS We were able to segregate 21 (68%) of 31 subjects into unlikely to respond categories based on Progression Free Survival at 6 months (PFS6) criteria. Twenty-two (69%) of 32 subjects could similarly be identified as unlikely to survive long using the machine learning algorithm. CONCLUSION With the use of machine learning techniques to evaluate imaging features derived from pre- and post-treatment multimodal MRI, it is possible to identify an important fraction of patients who are either highly unlikely to respond, or highly likely to respond. This can be helpful is selecting patients that either should or should not be treated with BV.
Collapse
Affiliation(s)
- Lucie Petrova
- Department of Anesthesiology and Critical Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria; Austria and Department of Neurosurgery, Military Hospital in Prague, 16902 Praha 6, Czech Republic
| | - Panagiotis Korfiatis
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Ondra Petr
- Department of Neurosurgery, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Daniel H LaChance
- Department of Neurology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Ian Parney
- Department of Neurosurgery, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Jan C Buckner
- Department of Oncology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America
| | - Bradley J Erickson
- Department of Radiology, Mayo Clinic, 200 First St SW, Rochester, MN 55905, United States of America.
| |
Collapse
|
15
|
Mendes M, Sousa JJ, Pais A, Vitorino C. Targeted Theranostic Nanoparticles for Brain Tumor Treatment. Pharmaceutics 2018; 10:E181. [PMID: 30304861 PMCID: PMC6321593 DOI: 10.3390/pharmaceutics10040181] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/13/2022] Open
Abstract
The poor prognosis and rapid recurrence of glioblastoma (GB) are associated to its fast-growing process and invasive nature, which make difficult the complete removal of the cancer infiltrated tissues. Additionally, GB heterogeneity within and between patients demands a patient-focused method of treatment. Thus, the implementation of nanotechnology is an attractive approach considering all anatomic issues of GB, since it will potentially improve brain drug distribution, due to the interaction between the blood⁻brain barrier and nanoparticles (NPs). In recent years, theranostic techniques have also been proposed and regarded as promising. NPs are advantageous for this application, due to their respective size, easy surface modification and versatility to integrate multiple functional components in one system. The design of nanoparticles focused on therapeutic and diagnostic applications has increased exponentially for the treatment of cancer. This dual approach helps to understand the location of the tumor tissue, the biodistribution of nanoparticles, the progress and efficacy of the treatment, and is highly useful for personalized medicine-based therapeutic interventions. To improve theranostic approaches, different active strategies can be used to modulate the surface of the nanotheranostic particle, including surface markers, proteins, drugs or genes, and take advantage of the characteristics of the microenvironment using stimuli responsive triggers. This review focuses on the different strategies to improve the GB treatment, describing some cell surface markers and their ligands, and reports some strategies, and their efficacy, used in the current research.
Collapse
Affiliation(s)
- Maria Mendes
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
| | - João José Sousa
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| | - Alberto Pais
- Coimbra Chemistry Centre, Department of Chemistry, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal.
- Center for Neurosciences and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal.
- LAQV, REQUIMTE, Group of Pharmaceutical Technology, 3000-548 Coimbra, Portugal.
| |
Collapse
|
16
|
Hong B, Polemikos M, Heissler HE, Hartmann C, Nakamura M, Krauss JK. Challenges in cerebrospinal fluid shunting in patients with glioblastoma. Fluids Barriers CNS 2018; 15:16. [PMID: 29860942 PMCID: PMC5985574 DOI: 10.1186/s12987-018-0101-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 01/17/2023] Open
Abstract
Background Cerebrospinal fluid (CSF) circulation disturbances may occur during the course of disease in patients with glioblastoma. Ventriculoperitoneal shunting has generally been recommended to improve symptoms in glioblastoma patients. Shunt implantation for patients with glioblastoma, however, presents as a complex situation and produces different problems to shunting in other contexts. Information on complications of shunting glioma patients has rarely been the subject of investigation. In this retrospective study, we analysed restropectively the course and outcome of glioblastoma-related CSF circulation disturbances after shunt management in a consecutive series of patients within a period of over a decade. Methods Thirty of 723 patients with histopathologically-confirmed glioblastoma diagnosed from 2002 to 2016 at the Department of Neurosurgery, Hannover Medical School, underwent shunting for CSF circulation disorders. Treatment history of glioblastoma and all procedures associated with shunt implementation were analyzed. Data on follow-up, time to progression and survival rates were obtained by review of hospital charts and supplemented by phone interviews with the patients, their relations or the primary physicians. Results Mean age at the time of diagnosis of glioblastoma was 43 years. Five types of CSF circulation disturbances were identified: obstructive hydrocephalus (n = 9), communicating hydrocephalus (n = 15), external hydrocephalus (n = 3), trapped lateral ventricle (n = 1), and expanding fluid collection in the resection cavity (n = 2). All patients showed clinical deterioration. Procedures for CSF diversion were ventriculoperitoneal shunt (n = 21), subduroperitoneal shunt (n = 3), and cystoperitoneal shunt (n = 2). In patients with lower Karnofsky Performance Score (KPS) (< 60), there was a significant improvement of median KPS after shunt implantation (p = 0.019). Shunt revision was necessary in 9 patients (single revision, n = 6; multiple revisions, n = 3) due to catheter obstruction, catheter dislocation, valve defect, and infection. Twenty-eight patients died due to disease progression during a median follow-up time of 88 months. The median overall survival time after diagnosis of glioblastoma was 10.18 months. Conclusions CSF shunting in glioblastoma patients encounters more challenge and is associated with increased risk of complications, but these can be usually managed by revision surgeries. CSF shunting improves neurological function temporarily, enhances quality of life in most patients although it is not known if survival rate is improved.
Collapse
Affiliation(s)
- Bujung Hong
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany.
| | - Manolis Polemikos
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Hans E Heissler
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| | - Christian Hartmann
- Institute for Pathology, Department for Neuropathology, Hannover Medical School, Hannover, Germany
| | - Makoto Nakamura
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany.,Department of Neurosurgery, Cologne Mehrheim Medical Center, University of Witten/Herdecke, Cologne, Germany
| | - Joachim K Krauss
- Department of Neurosurgery, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Kratzsch T, Kuhn SA, Joedicke A, Hanisch UK, Vajkoczy P, Hoffmann J, Fichtner I. Treatment with 5-azacitidine delay growth of glioblastoma xenografts: a potential new treatment approach for glioblastomas. J Cancer Res Clin Oncol 2018; 144:809-819. [PMID: 29427211 DOI: 10.1007/s00432-018-2600-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/30/2018] [Indexed: 12/31/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults. The epigenetically active ribonucleoside analog 5-azacitidine is a new therapy option that changes tumor cell chromatin, which is frequently modified by methylation and deacetylation in malignant gliomas. METHODS In vitro, we analyzed cell viability, cell apoptosis, and migration of human GBM cells. In vivo, we established subcutaneous and intracerebral GBM mouse models originating from U87MG, U373MG, and primary GBM cells as well as one patient-derived xenograft. Xenografts were treated with 5-azacitidine as well as valproic acid, bevacizumab, temozolomide, and phosphate buffered saline. The tumor sizes and Ki67 proliferation indices were determined. Glioma angiogenesis was examined immunohistochemically by expression analysis of endothelial cells (CD31) and pericytes (PDGFRβ). RESULTS In vitro, 5-azacitidine treatment significantly reduced human glioblastoma cell viability, increased cellular apoptosis, and reduced cellular migration. In vivo, 5-azacitidine significantly reduced growth in two intracerebral GBM models. Notably, this was also shown for a xenograft established from a patient surgery sample; whereas, epigenetically acting valproic acid did not show any growth reduction. Highly vascularized tumors responded to treatment, whereas low-vascularized xenografts showed no response. Furthermore, intracerebral glioblastomas treated with 5-azacitidine showed a clearly visible reduction of tumor angiogenesis and lower numbers of endothelial cells and tumor vessel pericytes. CONCLUSIONS Our data show significant growth inhibition as well as antiangiogenic effects in intracerebral as well as patient-derived GBM xenografts. This encourages to investigate in detail the multifactorial effects of 5-azacitidine on glioblastomas.
Collapse
Affiliation(s)
- Tobias Kratzsch
- Department of Neurosurgery, Charité University Hospital, Chariteplatz 1, 10117, Berlin, Germany.
| | - Susanne Antje Kuhn
- Department of Neurosurgery, Ernst von Bergmann Hospital, Potsdam, Germany
| | - Andreas Joedicke
- Department of Neurosurgery, Vivantes Hospital Berlin Neukölln, Berlin, Germany
| | - Uwe Karsten Hanisch
- Institute of Neuropathology, University Hospital, Göttingen, Germany
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University Hospital, Chariteplatz 1, 10117, Berlin, Germany
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology GmbH, Berlin, Germany
| | - Iduna Fichtner
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
18
|
Adhesion- and stress-related adaptation of glioma radiochemoresistance is circumvented by β1 integrin/JNK co-targeting. Oncotarget 2018; 8:49224-49237. [PMID: 28514757 PMCID: PMC5564763 DOI: 10.18632/oncotarget.17480] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Abstract
Resistance of cancer stem-like and cancer tumor bulk cells to radiochemotherapy and destructive infiltration of the brain fundamentally influence the treatment efficiency to cure of patients suffering from Glioblastoma (GBM). The interplay of adhesion and stress-related signaling and activation of bypass cascades that counteract therapeutic approaches remain to be identified in GBM cells. We here show that combined inhibition of the adhesion receptor β1 integrin and the stress-mediator c-Jun N-terminal kinase (JNK) induces radiosensitization and blocks invasion in stem-like and patient-derived GBM cultures as well as in GBM cell lines. In vivo, this treatment approach not only significantly delays tumor growth but also increases median survival of orthotopic, radiochemotherapy-treated GBM mice. Both, in vitro and in vivo, effects seen with β1 integrin/JNK co-inhibition are superior to the monotherapy. Mechanistically, the in vitro radiosensitization provoked by β1 integrin/JNK targeting is caused by defective DNA repair associated with chromatin changes, enhanced ATM phosphorylation and prolonged G2/M cell cycle arrest. Our findings identify a β1 integrin/JNK co-dependent bypass signaling for GBM therapy resistance, which might be therapeutically exploitable.
Collapse
|
19
|
Hellton KH, Hjort NL. Fridge: Focused fine-tuning of ridge regression for personalized predictions. Stat Med 2018; 37:1290-1303. [DOI: 10.1002/sim.7576] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 11/06/2022]
Affiliation(s)
| | - Nils Lid Hjort
- Department of Mathematics; University of Oslo; Oslo Norway
| |
Collapse
|
20
|
Lei X, Wang F, Ke Y, Wei D, Gu H, Zhang Z, Jiang L, Lv L, Lin J, Wang L. The role of antiangiogenic agents in the treatment of gastric cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2017; 96:e6301. [PMID: 28272258 PMCID: PMC5348206 DOI: 10.1097/md.0000000000006301] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The survival of advanced gastric cancer (GC) is dismal, and effects of antiangiogenic agents remain inconclusive. The purpose of this study is to assess combination of chemotherapy with antiangiogenic therapy versus traditional chemotherapy. METHODS To achieve the goal of scientific rigor, statistics from both referenced works and experiments were analyzed. We carefully searched for the referenced works by retrieving, as well as analyzing, literature databases for information on antiangiogenic therapy compared to other therapeutic approaches used to treat GC patients. Two groups were defined in the experiment: experimental and control groups. The experimental group was treated with antiangiogenic drug, and the control group was treated with standard chemotherapy or placebo. RESULTS The study included a total of 3240 participants. Overall, there was significant improvement in overall survival (hazard ratio [HR] = 0.78, 95% confidence interval [CI]: 0.67-0.91, P = 0.002), progression-free survival (HR 0.65, 95% CI: 0.52-0.81, P = 0.0002), objective response rate (risk ratio [RR] = 1.58, 95% CI: 1.33-1.88, P < 0.00001), and disease control rate (RR 2.44, 95% CI: 1.57-3.78, P < 0.0001) in the group with antiangiogenic drug versus the group with standard chemotherapy or placebo. Moreover, this new treatment approach showed tolerable toxicity. CONCLUSION This study confirms the superior efficacy of combination therapy with antiangiogenic agents in comparison to traditional chemotherapy regimens for patients with GC. Moreover, this new treatment approach showed tolerable toxicity. This meta-analysis provides important information for clinicians who are interested in using antiangiogenic therapies to treat GC patients.
Collapse
Affiliation(s)
| | - Feng Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, PR China
| | - Yang Ke
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| | - Dong Wei
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| | - Hou Gu
- Department of Medical Oncology
| | | | | | - Li Lv
- Department of Medical Oncology
| | - Jie Lin
- Department of Medical Oncology
| | - Lin Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| |
Collapse
|
21
|
Esposito G, Burgunder JM, Dunlop J, Gorwood P, Inamdar A, Pfister SM, Pochet R, van den Bent MJ, Van Hoylandt N, Weller M, Westphal M, Wick W, Nutt D. Gene-Tailored Treatments for Brain Disorders: Challenges and Opportunities. Public Health Genomics 2016; 19:170-7. [DOI: 10.1159/000446535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
22
|
Targeting CD146 with a 64Cu-labeled antibody enables in vivo immunoPET imaging of high-grade gliomas. Proc Natl Acad Sci U S A 2015; 112:E6525-34. [PMID: 26553993 DOI: 10.1073/pnas.1502648112] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Given the highly heterogeneous character of brain malignancies and the associated implication for its proper diagnosis and treatment, finding biomarkers that better characterize this disease from a molecular standpoint is imperative. In this study, we evaluated CD146 as a potential molecular target for diagnosis and targeted therapy of glioblastoma multiforme (GBM), the most common and lethal brain malignancy. YY146, an anti-CD146 monoclonal antibody, was generated and radiolabeled for noninvasive positron-emission tomography (PET) imaging of orthotopic GBM models. (64)Cu-labeled YY146 preferentially accumulated in the tumors of mice bearing U87MG xenografts, which allowed the acquisition of high-contrast PET images of small tumor nodules (∼ 2 mm). Additionally, we found that tumor uptake correlated with the levels of CD146 expression in a highly specific manner. We also explored the potential therapeutic effects of YY146 on the cancer stem cell (CSC) and epithelial-to-mesenchymal (EMT) properties of U87MG cells, demonstrating that YY146 can mitigate those aggressive phenotypes. Using YY146 as the primary antibody, we performed histological studies of World Health Organization (WHO) grades I through IV primary gliomas. The positive correlation found between CD146-positive staining and high tumor grade (χ(2) = 9.028; P = 0.029) concurred with the GBM data available in The Cancer Genome Atlas (TCGA) and validated the clinical value of YY146. In addition, we demonstrate that YY146 can be used to detect CD146 in various cancer cell lines and human resected tumor tissues of multiple other tumor types (gastric, ovarian, liver, and lung), indicating a broad applicability of YY146 in solid tumors.
Collapse
|
23
|
Elkamhawy A, Viswanath ANI, Pae AN, Kim HY, Heo JC, Park WK, Lee CO, Yang H, Kim KH, Nam DH, Seol HJ, Cho H, Roh EJ. Discovery of potent and selective cytotoxic activity of new quinazoline-ureas against TMZ-resistant glioblastoma multiforme (GBM). Eur J Med Chem 2015; 103:210-22. [DOI: 10.1016/j.ejmech.2015.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 07/18/2015] [Accepted: 08/01/2015] [Indexed: 01/24/2023]
|
24
|
ElBanan MG, Amer AM, Zinn PO, Colen RR. Imaging genomics of Glioblastoma: state of the art bridge between genomics and neuroradiology. Neuroimaging Clin N Am 2015; 25:141-53. [PMID: 25476518 DOI: 10.1016/j.nic.2014.09.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastoma (GBM) is the most common and most aggressive primary malignant tumor of the central nervous system. Recently, researchers concluded that the "one-size-fits-all" approach for treatment of GBM is no longer valid and research should be directed toward more personalized and patient-tailored treatment protocols. Identification of the molecular and genomic pathways underlying GBM is essential for achieving this personalized and targeted therapeutic approach. Imaging genomics represents a new era as a noninvasive surrogate for genomic and molecular profile identification. This article discusses the basics of imaging genomics of GBM, its role in treatment decision-making, and its future potential in noninvasive genomic identification.
Collapse
Affiliation(s)
- Mohamed G ElBanan
- Department of Diagnostic Radiology, MD Anderson Cancer Center, University of Texas, 1400 Pressler Street, Houston, TX 77030, USA
| | - Ahmed M Amer
- Department of Diagnostic Radiology, MD Anderson Cancer Center, University of Texas, 1400 Pressler Street, Houston, TX 77030, USA
| | - Pascal O Zinn
- Department of Neurosurgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Rivka R Colen
- Department of Diagnostic Radiology, MD Anderson Cancer Center, University of Texas, 1400 Pressler Street, Houston, TX 77030, USA.
| |
Collapse
|
25
|
Jakola AS, Sagberg LM, Gulati S, Solheim O. Perioperative quality of life in functionally dependent glioblastoma patients: A prospective study. Br J Neurosurg 2015; 29:843-9. [PMID: 26098603 DOI: 10.3109/02688697.2015.1054355] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Functionally dependent patients with glioblastoma have a poor prognosis which may in part be due to a negative treatment selection. Prospective data on patient-reported quality of life (QoL) following surgery, together with an updated survival analysis, are lacking with regard to functionally dependent glioblastoma patients. MATERIALS AND METHODS Adult patients (≥ 18 years) with a histologically confirmed primary glioblastoma with preoperative Karnofsky Performance Status (KPS) ≤ 60, who were treated between January 1, 2007 and March 30, 2014, were eligible for inclusion. EuroQol 5D (EQ-5D) was scored before surgery and at 4-6 weeks postoperatively. A control group of 20 independent (i.e., KPS ≥ 70) adult primary glioblastoma patients was constructed. RESULTS Among the 27 patients included, only 22 patients were willing to participate in QoL research, with complete QoL follow-up data obtained for 16. There were 22 resections and 5 biopsies. In resection cases, the median extent of resection was 93% (IQR 78-99), with gross-total resection achieved in 24%. In the 16 patients with complete QoL data, the EQ-5D index score increased from 0.34 ± 0.38 preoperatively to 0.45 ± 0.32 postoperatively (p = 0.30), with 3 patients that deteriorated in QoL following surgery. Median survival was 7.3 months (95%, CI: 4.6-9.9, n = 27), and the perioperative mortality was 7%. Treatment characteristics between dependent and independent patients were similar, as were the unfavorable outcomes defined as QoL reduction, QoL drop-out, and dead before QoL assessment (41% vs. 45%, p = 0.79). However, a difference was seen in 6-month survival (42% vs. 15%, p = 0.05). CONCLUSION In most functionally dependent patients with glioblastoma, cytoreductive surgery is possible, and improved or unchanged postoperative QoL may be seen in approximately half of the cases. This must be weighed against the risk of complications and the modest effect of cytoreductive surgery.
Collapse
Affiliation(s)
- Asgeir S Jakola
- a Department of Neurosurgery , St. Olavs University Hospital , Trondheim , Norway.,b Norwegian Advisory Unit for Ultrasound and Image-Guided Surgery, St. Olavs University Hospital , Trondheim , Norway.,c Department of Neurosurgery , Sahlgrenska University Hospital , Gothenburg , Sweden
| | - Lisa M Sagberg
- a Department of Neurosurgery , St. Olavs University Hospital , Trondheim , Norway.,b Norwegian Advisory Unit for Ultrasound and Image-Guided Surgery, St. Olavs University Hospital , Trondheim , Norway.,e Department of Neuroscience , Norwegian University of Science and Technology , Trondheim , Norway
| | - Sasha Gulati
- a Department of Neurosurgery , St. Olavs University Hospital , Trondheim , Norway.,d Norwegian Centre of Competence in Deep Brain Stimulation for Movement Disorders, St. Olavs University Hospital , Trondheim , Norway
| | - Ole Solheim
- a Department of Neurosurgery , St. Olavs University Hospital , Trondheim , Norway.,b Norwegian Advisory Unit for Ultrasound and Image-Guided Surgery, St. Olavs University Hospital , Trondheim , Norway.,e Department of Neuroscience , Norwegian University of Science and Technology , Trondheim , Norway
| |
Collapse
|
26
|
Van Gool SW. Brain Tumor Immunotherapy: What have We Learned so Far? Front Oncol 2015; 5:98. [PMID: 26137448 PMCID: PMC4470276 DOI: 10.3389/fonc.2015.00098] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 04/13/2015] [Indexed: 12/17/2022] Open
Abstract
High grade glioma is a rare brain cancer, incurable in spite of modern neurosurgery, radiotherapy, and chemotherapy. Novel approaches are in research, and immunotherapy emerges as a promising strategy. Clinical experiences with active specific immunotherapy demonstrate feasibility, safety and most importantly, but incompletely understood, prolonged long-term survival in a fraction of the patients. In relapsed patients, we developed an immunotherapy schedule and we categorized patients into clinically defined risk profiles. We learned how to combine immunotherapy with standard multimodal treatment strategies for newly diagnosed glioblastoma multiforme patients. The developmental program allows further improvements related to newest scientific insights. Finally, we developed a mode of care within academic centers to organize cell-based therapies for experimental clinical trials in a large number of patients.
Collapse
|
27
|
Malignant optic glioma – the spectrum of disease in a case series. Graefes Arch Clin Exp Ophthalmol 2015; 253:1187-94. [DOI: 10.1007/s00417-015-3045-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/24/2015] [Accepted: 04/29/2015] [Indexed: 10/23/2022] Open
|
28
|
Ening G, Osterheld F, Capper D, Schmieder K, Brenke C. Risk factors for glioblastoma therapy associated complications. Clin Neurol Neurosurg 2015; 134:55-9. [PMID: 25942630 DOI: 10.1016/j.clineuro.2015.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 12/24/2014] [Accepted: 01/03/2015] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Thromboembolic events, seizures, neurologic symptoms and adverse effects from corticosteroids and chemotherapies are frequent clinical complications seen in Glioblastoma (GB) patients. The exact impact these have on dismal patient outcome has not been fully elucidated. We aimed at assessing treatment associated complications, evaluating the impact on survival and defining risk factors. METHODS Two hundred and thirty three consecutive adult patients operated on for newly diagnosed GB at a single tertiary institution over a 5-year-period (2006-2011) were assessed. Demographic parameters (age, gender, comorbidity status quantified by the Charlson-comorbidity-index (CCI), functional status computed by the Karnofsky Performance Scale (KPS), tumor characteristics (size, location, IDH-1 mutation and MGMT-Promotor-methylation-status) and treatment parameters (volumetrically quantified extent of resection and adjuvant therapy) were retrospectively reviewed. Complications assessed were recorded as neurological (N), surgical (S) and medical (M). Independent risk factor analysis was performed by the univariate and multivariate logistic regression method. Survival analysis was plotted by the Kaplan-Meier-method, influence of complication occurrence was evaluated by the log-rank test. RESULTS One hundred and fifty nine (68.2%) patients had a total of 281 complications (90 N, 174 M and 17 S). Univariate analysis identified age (P=0.003), KPS<70 (P=0.002), CCI>3 (P=0.03), eloquent tumor location (P=0.001) and therapy other than the standard radio-chemotherapy with temozolomide therapy (P=0.034) as risk factors for complications. Multivariate analysis extracted the eloquent tumor location (P=0.007, odds ratio 1.94) as a significant predictor for complications. Having a complication significantly decreased patient survival (P=0.015). CONCLUSIONS Complications significantly decrease GB patient survival. Age, poor functional status, other than standard adjuvant therapy and eloquent tumor location proved as significant risk factors for encountering a therapy associated complication. Not extensive surgery or tumor size but surgery at eloquent locations impacts complication occurrence the strongest with a 2 fold increased complication occurrence risk.
Collapse
Affiliation(s)
- Genevieve Ening
- Department of Neurosurgery, Knappschafts-Krankenhaus Bochum-Langendreer, Ruhr-University of Bochum, In der Schornau 23-25, 44892 Bochum, Germany; Department of Neurosurgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany.
| | - Fransiska Osterheld
- Department of Neurosurgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany
| | - David Capper
- Department of Neuropathology, Institute of Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 224, 69120 Heidelberg, Germany; Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 224, 69120 Heidelberg, Germany
| | - Kirsten Schmieder
- Department of Neurosurgery, Knappschafts-Krankenhaus Bochum-Langendreer, Ruhr-University of Bochum, In der Schornau 23-25, 44892 Bochum, Germany; Department of Neurosurgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany
| | - Christopher Brenke
- Department of Neurosurgery, Knappschafts-Krankenhaus Bochum-Langendreer, Ruhr-University of Bochum, In der Schornau 23-25, 44892 Bochum, Germany; Department of Neurosurgery, University Medical Center Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1, 68167 Mannheim, Germany
| |
Collapse
|
29
|
Cherny NI, de Vries EGE, Emanuel L, Fallowfield L, Francis PA, Gabizon A, Piccart MJ, Sidransky D, Soussan-Gutman L, Tziraki C. Words matter: distinguishing "personalized medicine" and "biologically personalized therapeutics". J Natl Cancer Inst 2014; 106:dju321. [PMID: 25293984 PMCID: PMC4568994 DOI: 10.1093/jnci/dju321] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/30/2014] [Accepted: 08/28/2014] [Indexed: 12/20/2022] Open
Abstract
"Personalized medicine" has become a generic term referring to techniques that evaluate either the host or the disease to enhance the likelihood of beneficial patient outcomes from treatment interventions. There is, however, much more to personalization of care than just identifying the biotherapeutic strategy with the highest likelihood of benefit. In its new meaning, "personalized medicine" could overshadow the individually tailored, whole-person care that is at the bedrock of what people need and want when they are ill. Since names and definitional terms set the scope of the discourse, they have the power to define what personalized medicine includes or does not include, thus influencing the scope of the professional purview regarding the delivery of personalized care. Taxonomic accuracy is important in understanding the differences between therapeutic interventions that are distinguishable in their aims, indications, scope, benefits, and risks. In order to restore the due emphasis to the patient and his or her needs, we assert that it is necessary, albeit belated, to deconflate the contemporary term "personalized medicine" by taxonomizing this therapeutic strategy more accurately as "biologically personalized therapeutics" (BPT). The scope of truly personalized medicine and its relationship to biologically personalized therapeutics is described, emphasizing that the best of care must give due recognition and emphasis to both BPT and truly personalized medicine.
Collapse
Affiliation(s)
- Nathan I Cherny
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT).
| | - Elisabeth G E de Vries
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Linda Emanuel
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Lesley Fallowfield
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Prudence A Francis
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Alberto Gabizon
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Martine J Piccart
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - David Sidransky
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Lior Soussan-Gutman
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| | - Chariklia Tziraki
- Cancer Pain and Palliative Medicine Service, Department of Medical Oncology, Shaare Zedek Medical Center, Jerusalem, Israel (NIC); Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands (EGEdV); Kellog School of Management and Northwestern University Medical School, Chicago, IL (LE); Sussex Health Outcomes Research & Education in Cancer (SHORE-C),Brighton & Sussex Medical School, University of Sussex, Falmer, UK (LF); Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (PAF); Department of Oncology, Shaare Zedek Medical Center, and Hebrew University-School of Medicine, Jerusalem, Israel (AG); Université Libre de Bruxelles, Jules Bordet Institute, Brussels, Belgium (MJP); Department of Otolaryngology and Oncology, Johns Hopkins University, Baltimore, MD (DS); Oncotest/Verify, Teva Pharmaceutical Industries, Petach Tikva, Israel (LS-G); Melabev Community Elders Care Research Department, Jerusalem, Israel (CT)
| |
Collapse
|
30
|
Schindler G, Capper D, Korshunov A, Schmieder K, Brenke C. Spinal metastasis of gliosarcoma: Array-based comparative genomic hybridization for confirmation of metastatic spread. J Clin Neurosci 2014; 21:1945-50. [DOI: 10.1016/j.jocn.2014.03.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 03/19/2014] [Accepted: 03/29/2014] [Indexed: 10/25/2022]
|
31
|
Moeckel S, Meyer K, Leukel P, Heudorfer F, Seliger C, Stangl C, Bogdahn U, Proescholdt M, Brawanski A, Vollmann-Zwerenz A, Riemenschneider MJ, Bosserhoff AK, Spang R, Hau P. Response-predictive gene expression profiling of glioma progenitor cells in vitro. PLoS One 2014; 9:e108632. [PMID: 25268354 PMCID: PMC4182559 DOI: 10.1371/journal.pone.0108632] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 08/24/2014] [Indexed: 12/15/2022] Open
Abstract
Background High-grade gliomas are amongst the most deadly human tumors. Treatment results are disappointing. Still, in several trials around 20% of patients respond to therapy. To date, diagnostic strategies to identify patients that will profit from a specific therapy do not exist. Methods In this study, we used serum-free short-term treated in vitro cell cultures to predict treatment response in vitro. This approach allowed us (a) to enrich specimens for brain tumor initiating cells and (b) to confront cells with a therapeutic agent before expression profiling. Results As a proof of principle we analyzed gene expression in 18 short-term serum-free cultures of high-grade gliomas enhanced for brain tumor initiating cells (BTIC) before and after in vitro treatment with the tyrosine kinase inhibitor Sunitinib. Profiles from treated progenitor cells allowed to predict therapy-induced impairment of proliferation in vitro. Conclusion For the tyrosine kinase inhibitor Sunitinib used in this dataset, the approach revealed additional predictive information in comparison to the evaluation of classical signaling analysis.
Collapse
Affiliation(s)
- Sylvia Moeckel
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Katharina Meyer
- Institute for Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Petra Leukel
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Fabian Heudorfer
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Corinna Seliger
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Christina Stangl
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Ulrich Bogdahn
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Alexander Brawanski
- Department of Neurosurgery, University Hospital Regensburg, Regensburg, Germany
| | - Arabel Vollmann-Zwerenz
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
| | | | | | - Rainer Spang
- Institute for Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, University Hospital Regensburg, Regensburg, Germany
- * E-mail:
| |
Collapse
|
32
|
Keunen O, Taxt T, Grüner R, Lund-Johansen M, Tonn JC, Pavlin T, Bjerkvig R, Niclou SP, Thorsen F. Multimodal imaging of gliomas in the context of evolving cellular and molecular therapies. Adv Drug Deliv Rev 2014; 76:98-115. [PMID: 25078721 DOI: 10.1016/j.addr.2014.07.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 07/14/2014] [Accepted: 07/22/2014] [Indexed: 01/18/2023]
Abstract
The vast majority of malignant gliomas relapse after surgery and standard radio-chemotherapy. Novel molecular and cellular therapies are thus being developed, targeting specific aspects of tumor growth. While histopathology remains the gold standard for tumor classification, neuroimaging has over the years taken a central role in the diagnosis and treatment follow up of brain tumors. It is used to detect and localize lesions, define the target area for biopsies, plan surgical and radiation interventions and assess tumor progression and treatment outcome. In recent years the application of novel drugs including anti-angiogenic agents that affect the tumor vasculature, has drastically modulated the outcome of brain tumor imaging. To properly evaluate the effects of emerging experimental therapies and successfully support treatment decisions, neuroimaging will have to evolve. Multi-modal imaging systems with existing and new contrast agents, molecular tracers, technological advances and advanced data analysis can all contribute to the establishment of disease relevant biomarkers that will improve disease management and patient care. In this review, we address the challenges of glioma imaging in the context of novel molecular and cellular therapies, and take a prospective look at emerging experimental and pre-clinical imaging techniques that bear the promise of meeting these challenges.
Collapse
|
33
|
Price RL, Chiocca EA. Evolution of malignant glioma treatment: from chemotherapy to vaccines to viruses. Neurosurgery 2014; 61 Suppl 1:74-83. [PMID: 25032534 PMCID: PMC4104417 DOI: 10.1227/neu.0000000000000390] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Richard Lee Price
- Dardinger Neuro-oncology Center, Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Ennio Antonio Chiocca
- Harvey Cushing Neuro-oncology Laboratories, Harvard Institutes of Medicine, Department of Neurosurgery and Institute for the Neurosciences at the Brigham, Brigham and Women’s/Faulkner Hospital and Center for Neuro-oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Bian EB, Li J, He XJ, Zong G, Jiang T, Li J, Zhao B. Epigenetic modification in gliomas: role of the histone methyltransferase EZH2. Expert Opin Ther Targets 2014; 18:1197-206. [PMID: 25046371 DOI: 10.1517/14728222.2014.941807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Gliomas are characterized by increased anaplasia, malignization, proliferation and invasion. They exhibit high resistance to standard treatment with combinations of radiotherapy and chemotherapy. They are currently the most common primary malignancy tumors in the brain that is related to a high mortality rate. Recently, increasing evidence suggests that EZH2 is involved in a number of glioma cell processes, including proliferation, apoptosis, invasion and angiogenesis. AREAS COVERED In this review, we emphasize the role of EZH2 in gliomas. We also address that EZH2 interacting with DNA methylation mediates transcriptional repression of specific genes in gliomas, and the regulation of EZH2 by microRNAs in gliomas. EXPERT OPINION Although the exact role of EZH2 in gliomas has not been fully elucidated, to understand the role of EZH2 proteins in epigenetic modification will provide valuable insights into the causes of gliomas, and pave the way to the potential future applications of EZH2 in the treatment of gliomas.
Collapse
Affiliation(s)
- Er-Bao Bian
- The Second Affiliated Hospital of Anhui Medical University, Department of Neurosurgery , Hefei 230601 , China
| | | | | | | | | | | | | |
Collapse
|
35
|
Dickinson P. Advances in diagnostic and treatment modalities for intracranial tumors. J Vet Intern Med 2014; 28:1165-85. [PMID: 24814688 PMCID: PMC4857954 DOI: 10.1111/jvim.12370] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 02/24/2014] [Accepted: 03/25/2014] [Indexed: 12/23/2022] Open
Abstract
Intracranial neoplasia is a common clinical condition in domestic companion animals, particularly in dogs. Application of advances in standard diagnostic and therapeutic modalities together with a broad interest in the development of novel translational therapeutic strategies in dogs has resulted in clinically relevant improvements in outcome for many canine patients. This review highlights the status of current diagnostic and therapeutic approaches to intracranial neoplasia and areas of novel treatment currently in development.
Collapse
Affiliation(s)
- P.J. Dickinson
- Department of Surgical and Radiological SciencesSchool of Veterinary MedicineUniversity of California DavisDavisCA
| |
Collapse
|
36
|
Shazeeb MS, Gupta S, Bogdanov A. MR signal amplification for imaging of the mutant EGF receptor in orthotopic human glioma model. Mol Imaging Biol 2014; 15:675-84. [PMID: 23733229 DOI: 10.1007/s11307-013-0653-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE To investigate the potential of targeted MR signal amplification strategy for imaging of EGF receptor variant III (EGFRvIII) overexpression associated with the infiltrating margin of aggressive orthotopic brain tumors. PROCEDURES F(ab')2 fragments of humanized anti-EGFRvIII monoclonal antibody (EMD72000) were linked to deglycosylated horseradish peroxidase (HRP) and glucose oxidase (GOX). Detection of the F(ab')2 conjugate pair colocalization in vivo was enabled by a subsequent IV injection of a low molecular weight paramagnetic substrate of HRP, diTyr-GdDTPA. RESULTS The delivery of the targeted fragments to the tumor was validated using SPECT/CT imaging of radiolabeled anti-EGFRvIII F(ab')2 conjugates. Further, by using 3 T MRI, we observed time-dependent differences in tumor signal intensity and signal retention at the endpoint depending on whether or not the animals were pre-injected with the anti-EGFRvIII F(ab')2 conjugates. CONCLUSIONS Imaging of EGFRvIII expression in vivo was enabled by consecutive administration of targeted F(ab')2 conjugates and a paramagnetic substrate resulting in a tumor-specific receptor detection with high specificity and resolution.
Collapse
Affiliation(s)
- Mohammed Salman Shazeeb
- Department of Radiology, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA
| | | | | |
Collapse
|
37
|
Zheng Q, Han L, Dong Y, Tian J, Huang W, Liu Z, Jia X, Jiang T, Zhang J, Li X, Kang C, Ren H. JAK2/STAT3 targeted therapy suppresses tumor invasion via disruption of the EGFRvIII/JAK2/STAT3 axis and associated focal adhesion in EGFRvIII-expressing glioblastoma. Neuro Oncol 2014; 16:1229-43. [PMID: 24861878 DOI: 10.1093/neuonc/nou046] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND As a commonly mutated form of the epidermal growth factor receptor, EGFRvIII strongly promotes glioblastoma (GBM) tumor invasion and progression, but the mechanisms underlying this promotion are not fully understood. METHODS Through gene manipulation, we established EGFRvIII-, wild-type EGFR-, and vector-expressing GBM cells. We used cDNA microarrays, bioinformatics analysis, target-blocking migration and invasion assays, Western blotting, and an orthotopic U87MG GBM model to examine the phenotypic shifts and treatment effects of EGFRvIII expression in vitro and in vivo. Confocal imaging, co-immunoprecipitation, and siRNA assays detected the focal adhesion-associated complex and their relationships to the EGFRvIII/JAK2/STAT3 axis in GBM cells. RESULTS The activation of JAK2/STAT3 signaling is vital for promoting migration and invasion in EGFRvIII-GBM cells. AG490 or WP1066, the JAK2/STAT3 inhibitors, specifically destroyed EGFRvIII/JAK2/STAT3-related focal adhesions and depleted the activation of EGFR/Akt/FAK and JAK2/STAT3 signaling, thereby abolishing the ability of EGFRvIII-expressing GBM cells to migrate and invade. Furthermore, the RNAi silencing of JAK2 in EGFRvIII-expressing GBM cells significantly attenuated their ability to migrate and invade; however, as a result of a potential EGFRvIII-JAK2-STAT3 activation loop, neither EGFR nor STAT3 knockdown yielded the same effects. Moreover, AG490 or JAK2 gene knockdown greatly suppressed tumor invasion and progression in the U87MG-EGFRvIII orthotopic models. CONCLUSION Taken together, our data demonstrate that JAK2/STAT3 signaling is essential for EGFRvIII-driven migration and invasion by promoting focal adhesion and stabilizing the EGFRvIII/JAK2/STAT3 axis. Targeting JAK2/STAT3 therapy, such as AG490, may have potential clinical implications for the tailored treatment of GBM patients bearing EGFRvIII-positive tumors.
Collapse
Affiliation(s)
- Qifan Zheng
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Lei Han
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Yucui Dong
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Jing Tian
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Wei Huang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Zhaoyu Liu
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Xiuzhi Jia
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Tao Jiang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Jianning Zhang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Xia Li
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Chunsheng Kang
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| | - Huan Ren
- Department of Immunology, Harbin Medical University; Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin, China (Q.-F.Z., Y.-C.D., J.T., W.H., Z.-Y.L., X.-Z.J., H.R.); Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China (T.J.); College of Bioinformatics, Harbin Medical University, Harbin, China (X.L.); Department of Neurosurgery, Tianjin Medical University General Hospital; Laboratory of Neuro-Oncology, Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China (L.H., C.-S.K., J.-N.Z.); Chinese Glioma Cooperative Group (CGCG) (L.H., T.J., C.-S.K.)
| |
Collapse
|
38
|
Silencing of HIF-1α enhances the radiation sensitivity of human glioma growth in vitro and in vivo. Neuropharmacology 2014; 89:168-74. [PMID: 24859610 DOI: 10.1016/j.neuropharm.2014.05.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 05/07/2014] [Accepted: 05/07/2014] [Indexed: 11/20/2022]
Abstract
Gliomas are the leading cause of cancer-related mortality worldwide, and the incidence is increasing. Because gliomas often become resistant to radiation treatment, it is urgent to develop novel therapeutic methods that are more effective and less toxic than current therapies so as to enhance patient survival and quality of life. Effective enhancement of radiation therapy for gliomas in vivo and in vitro was observed upon silencing of hypoxia-inducible factor 1α (HIF-1α) with RNA interference; this enhancement was related to changes in the cell cycle and apoptosis that were accompanied by modulation of Cdc2, cyclin B1, and Bcl-2 expression. Our data suggest that HIF-1α silencing combined with radiation therapy will increase the therapeutic efficacy of glioma treatment via regulation of cell cycle and apoptosis-related signaling pathways.
Collapse
|
39
|
Combined PDK1 and CHK1 inhibition is required to kill glioblastoma stem-like cells in vitro and in vivo. Cell Death Dis 2014; 5:e1223. [PMID: 24810059 PMCID: PMC4047898 DOI: 10.1038/cddis.2014.188] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 01/13/2023]
Abstract
Glioblastoma (GBM) is the most common and deadly adult brain tumor. Despite aggressive surgery, radiation, and chemotherapy, the life expectancy of patients diagnosed with GBM is ∼14 months. The extremely aggressive nature of GBM results from glioblastoma stem-like cells (GSCs) that sustain GBM growth, survive intensive chemotherapy, and give rise to tumor recurrence. There is accumulating evidence revealing that GSC resilience is because of concomitant activation of multiple survival pathways. In order to decode the signal transduction networks responsible for the malignant properties of GSCs, we analyzed a collection of GSC lines using a dual, but complementary, experimental approach, that is, reverse-phase protein microarrays (RPPMs) and kinase inhibitor library screening. We treated GSCs in vitro with clinically relevant concentrations of temozolomide (TMZ) and performed RPPM to detect changes in phosphorylation patterns that could be associated with resistance. In addition, we screened GSCs in vitro with a library of protein and lipid kinase inhibitors to identify specific targets involved in GSC survival and proliferation. We show that GSCs are relatively insensitive to TMZ treatment in terms of pathway activation and, although displaying heterogeneous individual phospho-proteomic profiles, most GSCs are resistant to specific inhibition of the major signaling pathways involved in cell survival and proliferation. However, simultaneous multipathway inhibition by the staurosporin derivative UCN-01 results in remarkable inhibition of GSC growth in vitro. The activity of UCN-01 on GSCs was confirmed in two in vivo models of GBM growth. Finally, we used RPPM to study the molecular and functional effects of UCN-01 and demonstrated that the sensitivity to UCN-01 correlates with activation of survival signals mediated by PDK1 and the DNA damage response initiated by CHK1. Taken together, our results suggest that a combined inhibition of PDK1 and CHK1 represents a potentially effective therapeutic approach to reduce the growth of human GBM.
Collapse
|
40
|
Multiscale design of cell-type-specific pharmacokinetic/pharmacodynamic models for personalized medicine: application to temozolomide in brain tumors. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e112. [PMID: 24785551 PMCID: PMC4017092 DOI: 10.1038/psp.2014.9] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/18/2014] [Indexed: 12/31/2022]
Abstract
Optimizing anticancer therapeutics needs to account for variable drug responses in heterogeneous cell populations within the tumor as well as in organs of toxicity. To address cell heterogeneity, we propose a multiscale modeling approach—from in vitro to preclinical and clinical studies—to develop cell-type–specific pharmacokinetic–pharmacodynamic (PK-PD) models. A physiologically based mechanistic modeling approach integrating data from aqueous solutions, U87 glioma cells, mice, and cancer patients was utilized to characterize the brain disposition of temozolomide (TMZ), the cornerstone of chemotherapy against glioblastoma multiforme. The final model represented intracellular normal brain and brain tumor compartments in which TMZ pH-dependent conversion to the DNA-alkylating species leads to the formation of DNA adducts that serve as an entry point for a PD model. This multiscale protocol can be extended to account for TMZ PK-PD in different cell populations, thus providing a critical tool to personalize TMZ-based chemotherapy on a cell-type–specific basis.
Collapse
|
41
|
Hydrocephalus after resection and adjuvant radiochemotherapy in patients with glioblastoma. Clin Neurol Neurosurg 2014; 120:27-31. [PMID: 24731571 DOI: 10.1016/j.clineuro.2014.02.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/05/2014] [Accepted: 02/17/2014] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Glioblastomas are the most common primary malignant brain tumors in adults with a poor prognosis. The current study sought to identify risk factors in glioblastoma patients that are closely associated with communicating hydrocephalus. METHODS We retrospectively analyzed data from 151 patients who were diagnosed with a glioblastoma between 2007 and 2011 and underwent complete surgical resection closely followed by adjuvant radiochemotherapy. RESULTS We observed a significant tendency toward communicating hydrocephalus in cases of ventricular opening during surgical tumor resection (Fisher's exact test p<0.001) and a noticeable, although not statistically significant, correlation between the onset of communicating hydrocephalus and evidence of leptomeningeal tumor dissemination (Fisher's exact test p=0.067). Additionally, there was a trend toward frontal tumor location and a larger tumor volume in patients with communicating hydrocephalus. The majority of patients suffering from communicating hydrocephalus received a cerebrospinal fluid (CSF) shunt implantation after radiation therapy (63.6%, Fisher's exact test p=0.000). CONCLUSION We identified the following risk factors associated with the onset of communicating hydrocephalus in glioblastoma patients: ventricular opening during tumor resection and leptomeningeal tumor dissemination. Shunt implantation seems to be safe and effective in these patients.
Collapse
|
42
|
Happold C, Roth P, Silginer M, Florea AM, Lamszus K, Frei K, Deenen R, Reifenberger G, Weller M. Interferon-β Induces Loss of Spherogenicity and Overcomes Therapy Resistance of Glioblastoma Stem Cells. Mol Cancer Ther 2014; 13:948-61. [DOI: 10.1158/1535-7163.mct-13-0772] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
43
|
Abstract
Advances in our understanding of glioma biology has led to an increase in targeted therapies in preclinical and clinical trials; however, cellular heterogeneity often precludes the targeted molecules from being found on all glioma cells, thus reducing the efficacy of these treatments. In contrast, one trait shared by virtually all tumor cells is altered (dysregulated) metabolism. Tumor cells have an increased reliance on glucose, suggesting that treatments affecting cellular metabolism may be an effective method to improve current therapies. Indeed, metabolism has been a focus of cancer research in the last few years, as many pathways long associated with tumor growth have been found to intersect metabolic pathways in the cell. The ketogenic diet (high fat, low carbohydrate and protein), caloric restriction, and fasting all cause a metabolic change, specifically, a reduction in blood glucose and an increase in blood ketones. We, and others, have demonstrated that these metabolic changes improve survival in animal models of malignant gliomas and can potentiate the anti-tumor effect of chemotherapies and radiation treatment. In this review we discuss the use of metabolic alteration for the treatment of malignant brain tumors.
Collapse
Affiliation(s)
- Eric C Woolf
- Neuro-Oncology Research, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013
| | - Adrienne C Scheck
- Neuro-Oncology Research, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ 85013
| |
Collapse
|
44
|
Nakada M, Kita D, Watanabe T, Hayashi Y, Hamada JI. The mechanism of chemoresistance against tyrosine kinase inhibitors in malignant glioma. Brain Tumor Pathol 2014; 31:198-207. [DOI: 10.1007/s10014-013-0174-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/27/2013] [Indexed: 11/24/2022]
|
45
|
Borrmann N, Friedrich S, Schwabe K, Hedrich HJ, Krauss JK, Knapp WH, Nakamura M, Meyer GJ, Walte A. Systemic treatment with 4-211Atphenylalanine enhances survival of rats with intracranial glioblastoma. Nuklearmedizin 2013; 52:212-21. [PMID: 24036694 DOI: 10.3413/nukmed-0580-13-05] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 08/22/2013] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Increased amino acid transport in brain tumours is used for diagnostic purposes. It has been shown that the α-emitting radionuclide astatine-211 labeled to L-phenylalanine is taken up by glioblastoma cells. We here tested, if systemic treatment with 4-[211At]astatine-phenylalanine (At-Phe) has a beneficial effect on survival of rats with intracranial glioblastoma. ANIMALS, METHODS The rat glioblastoma cell line BT4Ca was implanted into the prefrontal cortex of female BDIX rats by stereotaxic microinjection (10,000 cells/3 µl; n = 83). 3 days after implantation At-Phe or phosphate buffered saline were injected intravenously. A third group was treated twice, i.e., on day 3 and 10. Health condition was assessed each day by using a score system. Rats were sacrificed on days 6, 10, 13 and 17 after implantation, or when showing premortal health condition to measure tumour volume and necrosis. The proliferation index (PI) was assessed after immunohistochemical staining of Ki-67. RESULTS Survival time of rats treated twice with At-Phe was significantly prolonged. Additionally, both At-Phe-treated groups remained significantly longer in a better health condition. Rats with poor health status had larger tumours than rats with fair health condition. Overall, irrespective of treatment the PI was reduced in rats with poor health condition. Necrosis was larger in rats treated twice with At-Phe. CONCLUSION Intravenous treatment with At-Phe enhanced survival time of rats with intracranial glioblastomas and improved health condition. These results encourage studies using local treatment of intracranial glioblastoma with At-Phe, either by repeated local injection or by intracavital application after tumour resection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - A Walte
- Dr. Almut Walte, Klinik für Nuklearmedizin, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1 30625 Hannover, Germany, Tel. +49/(0)511/532 40 11, E-mail:
| |
Collapse
|
46
|
Cloughesy TF, Cavenee WK, Mischel PS. Glioblastoma: from molecular pathology to targeted treatment. ANNUAL REVIEW OF PATHOLOGY 2013; 9:1-25. [PMID: 23937436 DOI: 10.1146/annurev-pathol-011110-130324] [Citation(s) in RCA: 409] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Glioblastoma (GBM) is one of the most lethal human cancers. Genomic analyses are defining the molecular architecture of GBM, uncovering relevant subsets of patients whose disease may require different treatments. Many pharmacological targets have been revealed, promising to transform patient care through targeted therapies. However, for most patients, clinical responses to targeted inhibitors are either not apparent or not durable. In this review, we address the challenge of developing more effective, molecularly guided approaches for the treatment of GBM patients. We summarize the current state of knowledge regarding molecular classifiers and examine their benefit for stratifying patients for treatment. We survey the molecular landscape of the disease, discussing the challenges raised by acquired drug resistance. Furthermore, we analyze the biochemical features of GBM, suggesting a next generation of drug targets, and we examine the contribution of tumor heterogeneity and its implications. We conclude with an analysis of the experimental approaches and their potential benefit to patients.
Collapse
Affiliation(s)
- Timothy F Cloughesy
- Department of Neurology and Neuro-Oncology Program, University of California, Los Angeles, California 90095;
| | | | | |
Collapse
|
47
|
Vehlow A, Cordes N. Invasion as target for therapy of glioblastoma multiforme. Biochim Biophys Acta Rev Cancer 2013; 1836:236-44. [PMID: 23891970 DOI: 10.1016/j.bbcan.2013.07.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 12/27/2022]
Abstract
The survival of cancer patients suffering from glioblastoma multiforme is limited to just a few months even after treatment with the most advanced techniques. The indefinable borders of glioblastoma cell infiltration into the surrounding healthy tissue prevent complete surgical removal. In addition, genetic mutations, epigenetic modifications and microenvironmental heterogeneity cause resistance to radio- and chemotherapy altogether resulting in a hardly to overcome therapeutic scenario. Therefore, the development of efficient therapeutic strategies to combat these tumors requires a better knowledge of genetic and proteomic alterations as well as the infiltrative behavior of glioblastoma cells and how this can be targeted. Among many cell surface receptors, members of the integrin family are known to regulate glioblastoma cell invasion in concert with extracellular matrix degrading proteases. While preclinical and early clinical trials suggested specific integrin targeting as a promising therapeutic approach, clinical trials failed to deliver improved cure rates up to now. Little is known about glioblastoma cell motility, but switches in invasion modes and adaption to specific microenvironmental cues as a consequence of treatment may maintain tumor cell resistance to therapy. Thus, understanding the molecular basis of integrin and protease function for glioblastoma cell invasion in the context of radiochemotherapy is a pressing issue and may be beneficial for the design of efficient therapeutic approaches. This review article summarizes the latest findings on integrins and extracellular matrix in glioblastoma and adds some perspective thoughts on how this knowledge might be exploited for optimized multimodal therapy approaches.
Collapse
Affiliation(s)
- Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, 01307 Dresden, Germany
| | | |
Collapse
|
48
|
Chang HT, Olson LK, Schwartz KA. Ketolytic and glycolytic enzymatic expression profiles in malignant gliomas: implication for ketogenic diet therapy. Nutr Metab (Lond) 2013; 10:47. [PMID: 23829383 PMCID: PMC3707813 DOI: 10.1186/1743-7075-10-47] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/27/2013] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Recent studies in animal models, based on the hypothesis that malignant glioma cells are more dependent on glycolysis for energy generation, have shown promising results using ketogenic diet (KD) therapy as an alternative treatment strategy for malignant glioma, effectively starving glioma cells while providing ketone bodies as an energy source for normal neurons and glial cells. In order to test this treatment strategy in humans, we investigated the relative expression of several key enzymes involved in ketolytic and glycolytic metabolism in human anaplastic glioma (WHO grade III) and glioblastoma (GBM, WHO grade IV). METHODS Immunohistochemistry was performed on formalin fixed paraffin embedded sections from 22 brain biopsies (17 GBM, 3 anaplastic astrocytoma and 2 anaplastic oligoastrocytoma) using antibodies raised against glycolytic and ketolytic enzymes. The glycolytic enzymes included hexokinase-II (HK2) and pyruvate kinase M2 isoform (PKM2). The ketone body metabolic enzymes included: succinyl CoA: 3-oxoacid CoA transferase (OXCT1), 3-hydroxybutyrate dehydrogenase 1 and 2 (BDH1 and BDH2), and acetyl-CoA acetyltransferase 1 (ACAT1). The immunoreactivities were graded using a semi-quantitative scale based on the percentage of positive cells: POS (>20%), LOW (5-20%), and very low (VLOW) (<5%). Focal non-neoplastic "normal" brain tissue within the biopsy specimens served as internal controls. RESULTS The rate limiting mitochondrial ketolytic enzymes (OXCT1 and BDH1) were either LOW or VLOW, concordantly in 14 of the 17 GBMs and in 1 of 5 anaplastic gliomas, whereas at least one of the glycolytic enzymes was POS in 13 of these 17 GBMs and all 5 anaplastic gliomas. Cytosolic BDH2 and mitochondrial ACTAT1 were, surprisingly, POS in most of these tumors. CONCLUSION Our results showing that malignant gliomas have differential expression of ketolytic and glycolytic enzymes are consistent with previous studies that have shown that these are genetically heterogeneous tumors. It seems reasonable to hypothesize that patients with low or very low expression of key ketolytic enzymes in their malignant gliomas may respond better to the KD therapy than those patients with positive expression of these enzymes. Further studies in animal models and/or a large-scale clinical trial would be needed to test this hypothesis.
Collapse
Affiliation(s)
- Howard T Chang
- Department of Neurology and Ophthalmology, Michigan State University, East Lansing, MI, 48824, USA.
| | | | | |
Collapse
|
49
|
Zhang XQ, Sun S, Lam KF, Kiang KMY, Pu JKS, Ho ASW, Lui WM, Fung CF, Wong TS, Leung GKK. A long non-coding RNA signature in glioblastoma multiforme predicts survival. Neurobiol Dis 2013; 58:123-31. [PMID: 23726844 DOI: 10.1016/j.nbd.2013.05.011] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/08/2013] [Accepted: 05/20/2013] [Indexed: 12/19/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) represent the leading edge of cancer research, and have been implicated in cancer biogenesis and prognosis. We aimed to identify lncRNA signatures that have prognostic values in glioblastoma multiforme (GBM). Using a lncRNA-mining approach, we performed lncRNA expression profiling in 213 GBM tumors from The Cancer Genome Atlas (TCGA), randomly divided into a training (n=107) and a testing set (n=106). We analyzed the associations between lncRNA signatures and clinical outcome in the training set, and validated the findings in the testing set. We also validated the identified lncRNA signature in another two independent GBM data sets from Gene Expression Omnibus (GEO), which contained specimens from 68 and 101 patients, respectively. We identified a set of six lncRNAs that were significantly associated with the overall survival in the training set (P≤0.01). Based on this six-lncRNA signature, the training-set patients could be classified into high-risk and low-risk subgroups with significantly different survival (HR=2.13, 95% CI=1.38-3.29; P=0.001). The prognostic value of this six-lncRNA signature was confirmed in the testing set and the two independent data sets. Further analysis revealed that the prognostic value of this signature was independent of age and O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation status. The identification of the prognostic lncRNAs indicates the potential roles of lncRNAs in GBM pathogenesis. This six-lncRNA signature may have clinical implications in the subclassification of GBM.
Collapse
Affiliation(s)
- Xiao-Qin Zhang
- Department of Surgery, The University of Hong Kong, Hong Kong
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Using a preclinical mouse model of high-grade astrocytoma to optimize p53 restoration therapy. Proc Natl Acad Sci U S A 2013; 110:E1480-9. [PMID: 23542378 DOI: 10.1073/pnas.1219142110] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Based on clinical presentation, glioblastoma (GBM) is stratified into primary and secondary types. The protein 53 (p53) pathway is functionally incapacitated in most GBMs by distinctive type-specific mechanisms. To model human gliomagenesis, we used a GFAP-HRas(V12) mouse model crossed into the p53ER(TAM) background, such that either one or both copies of endogenous p53 is replaced by a conditional p53ER(TAM) allele. The p53ER(TAM) protein can be toggled reversibly in vivo between wild-type and inactive conformations by administration or withdrawal of 4-hydroxytamoxifen (4-OHT), respectively. Surprisingly, gliomas that develop in GFAP-HRas(V12);p53(+/KI) mice abrogate the p53 pathway by mutating p19(ARF)/MDM2 while retaining wild-type p53 allele. Consequently, such tumors are unaffected by restoration of their p53ER(TAM) allele. By contrast, gliomas arising in GFAP-HRas(V12);p53(KI/KI) mice develop in the absence of functional p53. Such tumors retain a functional p19(ARF)/MDM2-signaling pathway, and restoration of p53ER(TAM) allele triggers p53-tumor-suppressor activity. Congruently, growth inhibition upon normalization of mutant p53 by a small molecule, Prima-1, in human GBM cultures also requires p14(ARF)/MDM2 functionality. Notably, the antitumoral efficacy of p53 restoration in tumor-bearing GFAP-HRas(V12);p53(KI/KI) animals depends on the duration and frequency of p53 restoration. Thus, intermittent exposure to p53ER(TAM) activity mitigated the selective pressure to inactivate the p19(ARF)/MDM2/p53 pathway as a means of resistance, extending progression-free survival. Our results suggest that intermittent dosing regimes of drugs that restore wild-type tumor-suppressor function onto mutant, inactive p53 proteins will prove to be more efficacious than traditional chronic dosing by similarly reducing adaptive resistance.
Collapse
|