1
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15. [DOI: https:/doi.org/10.3389/fnmol.2022.910543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
|
2
|
Wu H, Wei M, Li Y, Ma Q, Zhang H. Research Progress on the Regulation Mechanism of Key Signal Pathways Affecting the Prognosis of Glioma. Front Mol Neurosci 2022; 15:910543. [PMID: 35935338 PMCID: PMC9354928 DOI: 10.3389/fnmol.2022.910543] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
As is known to all, glioma, a global difficult problem, has a high malignant degree, high recurrence rate and poor prognosis. We analyzed and summarized signal pathway of the Hippo/YAP, PI3K/AKT/mTOR, miRNA, WNT/β-catenin, Notch, Hedgehog, TGF-β, TCS/mTORC1 signal pathway, JAK/STAT signal pathway, MAPK signaling pathway, the relationship between BBB and signal pathways and the mechanism of key enzymes in glioma. It is concluded that Yap1 inhibitor may become an effective target for the treatment of glioma in the near future through efforts of generation after generation. Inhibiting PI3K/Akt/mTOR, Shh, Wnt/β-Catenin, and HIF-1α can reduce the migration ability and drug resistance of tumor cells to improve the prognosis of glioma. The analysis shows that Notch1 and Sox2 have a positive feedback regulation mechanism, and Notch4 predicts the malignant degree of glioma. In this way, notch cannot only be treated for glioma stem cells in clinic, but also be used as an evaluation index to evaluate the prognosis, and provide an exploratory attempt for the direction of glioma treatment. MiRNA plays an important role in diagnosis, and in the treatment of glioma, VPS25, KCNQ1OT1, KB-1460A1.5, and CKAP4 are promising prognostic indicators and a potential therapeutic targets for glioma, meanwhile, Rheb is also a potent activator of Signaling cross-talk etc. It is believed that these studies will help us to have a deeper understanding of glioma, so that we will find new and better treatment schemes to gradually conquer the problem of glioma.
Collapse
Affiliation(s)
- Hao Wu
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Min Wei
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Yuping Li
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Qiang Ma
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| | - Hengzhu Zhang
- Graduate School of Dalian Medical University, Dalian, China
- Department of Neurosurgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Dalian, China
| |
Collapse
|
3
|
Nguyen T, Mueller S, Malbari F. Review: Neurological Complications From Therapies for Pediatric Brain Tumors. Front Oncol 2022; 12:853034. [PMID: 35480100 PMCID: PMC9035987 DOI: 10.3389/fonc.2022.853034] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022] Open
Abstract
Surgery, chemotherapy and radiation have been the mainstay of pediatric brain tumor treatment over the past decades. Recently, new treatment modalities have emerged for the management of pediatric brain tumors. These therapies range from novel radiotherapy techniques and targeted immunotherapies to checkpoint inhibitors and T cell transfer therapies. These treatments are currently investigated with the goal of improving survival and decreasing morbidity. However, compared to traditional therapies, these novel modalities are not as well elucidated and similarly has the potential to cause significant short and long-term sequelae, impacting quality of life. Treatment complications are commonly mediated through direct drug toxicity or vascular, infectious, or autoimmune mechanisms, ranging from immune effector cell associated neurotoxicity syndrome with CART-cells to neuropathy with checkpoint inhibitors. Addressing treatment-induced complications is the focus of new trials, specifically improving neurocognitive outcomes. The aim of this review is to explore the pathophysiology underlying treatment related neurologic side effects, highlight associated complications, and describe the future direction of brain tumor protocols. Increasing awareness of these neurologic complications from novel therapies underscores the need for quality-of-life metrics and considerations in clinical trials to decrease associated treatment-induced morbidity.
Collapse
Affiliation(s)
- Thien Nguyen
- Department of Pediatrics, University of San Francisco, San Francisco, CA, United States
- *Correspondence: Thien Nguyen,
| | - Sabine Mueller
- Department of Neurology, Neurosurgery and Pediatrics, University of San Francisco, San Francisco, CA, United States
| | - Fatema Malbari
- Division of Neurology, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Cabral de Carvalho Corrêa D, Tesser-Gamba F, Dias Oliveira I, Saba da Silva N, Capellano AM, de Seixas Alves MT, Dastoli PA, Cavalheiro S, Caminada de Toledo SR. Gliomas in children and adolescents: investigation of molecular alterations with a potential prognostic and therapeutic impact. J Cancer Res Clin Oncol 2021; 148:107-119. [PMID: 34626238 DOI: 10.1007/s00432-021-03813-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/21/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Gliomas represent the most frequent central nervous system (CNS) tumors in children and adolescents. However, therapeutic strategies for these patients, based on tumor molecular profile, are still limited compared to the wide range of treatment options for the adult population. We investigated molecular alterations, with a potential prognostic marker and therapeutic target in gliomas of childhood and adolescence using the next-generation sequencing (NGS) strategy. METHODS We selected 95 samples with initial diagnosis of glioma from patients treated at Pediatric Oncology Institute-GRAACC/UNIFESP. All samples were categorized according to the 2021 World Health Organization Classification of Tumors of the CNS, which included 39 low-grade gliomas (LGGs) and 56 high-grade gliomas (HGGs). Four HGG samples were classified as congenital glioblastoma (cGBM). NGS was performed to identify somatic genetic variants in tumor samples using the Oncomine Childhood Cancer Research Assay® (OCCRA®) panel, from Thermo Fisher Scientific®. RESULTS Genetic variants were identified in 76 of 95 (80%) tumors. In HGGs, the most common molecular alteration detected was H3F3A c.83A > T variant (H3.3 K27M) and co-occurring mutations in ATRX, TP53, PDGFRA, MET, and MYC genes were also frequently observed. One HGG sample was reclassified as supratentorial ependymoma ZFTA-fusion positive after NGS was performed. In LGGs, four KIAA1549-BRAF fusion transcripts were detected and this alteration was the most recurrent genetic event and favorable prognostic factor identified. Additionally, genetic variants in ALK and NTRK genes, which provide potential targets for therapy with Food and Drug Administration-approved drugs, were identified in two different cases of cGBM that were classified as infant-type hemispheric glioma, a newly recognized subgroup of pediatric HGG. CONCLUSION Molecular profiling by the OCCRA® panel comprehensively addressed the most relevant genetic variants in gliomas of childhood and adolescence, as these tumors have specific patterns of molecular alterations, outcomes, and effectiveness to therapies.
Collapse
Affiliation(s)
- Débora Cabral de Carvalho Corrêa
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil.,Division of Genetics, Department of Morphology and Genetics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Francine Tesser-Gamba
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil
| | - Indhira Dias Oliveira
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil
| | - Nasjla Saba da Silva
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil
| | - Andrea Maria Capellano
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil
| | - Maria Teresa de Seixas Alves
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil.,Department of Pathology, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Patrícia Alessandra Dastoli
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil.,Department of Neurology and Neurosurgery, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Sergio Cavalheiro
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil.,Department of Neurology and Neurosurgery, Federal University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Silvia Regina Caminada de Toledo
- Department of Pediatrics, Pediatric Oncology Institute-GRAACC/UNIFESP, Federal University of Sao Paulo, 743 Botucatu Street, 8th Floor - Genetics Laboratory, Vila Clementino, Sao Paulo, SP, 04023-062, Brazil. .,Division of Genetics, Department of Morphology and Genetics, Federal University of Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
5
|
Wu M, Hu W, Wang G, Yao Y, Yu XF. Nicotinamide N-Methyltransferase Is a Prognostic Biomarker and Correlated With Immune Infiltrates in Gastric Cancer. Front Genet 2020; 11:580299. [PMID: 33193702 PMCID: PMC7655872 DOI: 10.3389/fgene.2020.580299] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the third most common cause of cancer-related death in the word. Immunotherapy is a promising treatment of cancer. However, it is unclear which GC subpopulation would benefit most from immunotherapy and it is necessary to develop effective biomarkers for predicting immunotherapy response. Nicotinamide N-methyltransferase (NNMT) is a metabolic regulator of cancer-associated fibroblast (CAF) differentiation and cancer progression. In this study, we explored the correlations of NNMT to tumor-infiltrating immune cells (TIICs) and immune marker sets in The Cancer Genome Atlas Stomach Adenocarcinoma STAD (TCGA-STAD). Subsequently, we screened the NNMT correlated genes and performed the enrichment analysis of these genes. We eventually predicted the 19 most potential small-molecule drugs using the connectivity map (CMap) and Comparative Toxicogenomics Database (CTD). Also, nadolol, tranexamic acid, felbinac and dapsone were considered the four most promising drugs for GC. In summary, NNMT can be used as a prognostic biomarker that reflect immune infiltration level and a novel therapeutic target in GC.
Collapse
Affiliation(s)
- Miaowei Wu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| | - Weilei Hu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Guosheng Wang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihan Yao
- Department of Surgical Oncology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Fang Yu
- Cancer Institute, Second Affiliated Hospital, Cancer Institute, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Gao H, Chu C, Cheng Y, Zhang Y, Pang X, Li D, Wang X, Ren E, Xie F, Bai Y, Chen L, Liu G, Wang M. In Situ Formation of Nanotheranostics to Overcome the Blood-Brain Barrier and Enhance Treatment of Orthotopic Glioma. ACS APPLIED MATERIALS & INTERFACES 2020; 12:26880-26892. [PMID: 32441504 DOI: 10.1021/acsami.0c03873] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Glioblastoma is one of the most lethal cancers and needs effective therapeutics. The development of coordination-driven metal-organic nanoassemblies, which can cross the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) and have multiple desired functions, may provide a promising solution to this issue. Here, we report an in situ assembled nanoplatform based on RGD peptide-modified bisulfite-zincII-dipicolylamine-Arg-Gly-Asp (Bis(DPA-Zn)-RGD) and ultrasmall Au-ICG nanoparticles. Attributed to its positive charges and neovascular targeting properties, Bis(DPA-Zn)-RGD can be selectively delivered to the tumor site, and then assembled in situ into large nanoclusters with subsequently administered Au-ICG nanoparticles. Au nanoparticles with ultrasmall size (∼7 nm) can successfully cross the BBB. The obtained nanoclusters exhibit strong near-infrared-red (NIR) absorption and an enhanced tumor retention effect, enabling precise orthotopic fluorescence/photoacoustic imaging. With the aid of image guidance, the photothermal effect of the nanoclusters is observed to suppress tumor progression with the inhibition efficiency reaching up to 93.9%. Meanwhile, no photothermal damage can be found for normal brain tissues. These results, herein, suggest a feasible nanotheranostic agent with the ability to overcome the BBB and BBTB for imaging and therapy of orthotopic brain tumors.
Collapse
Affiliation(s)
- Haiyan Gao
- Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou 450003, P. R. China
- Henan Key Laboratory of Neurological Imaging, Zhengzhou University, Zhengzhou 450003, P. R. China
| | - Chengchao Chu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Yi Cheng
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Yang Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Dengfeng Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - En Ren
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Fengfei Xie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Yan Bai
- Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou 450003, P. R. China
- Henan Key Laboratory of Neurological Imaging, Zhengzhou University, Zhengzhou 450003, P. R. China
| | - Lijuan Chen
- Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou 450003, P. R. China
- Henan Key Laboratory of Neurological Imaging, Zhengzhou University, Zhengzhou 450003, P. R. China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Meiyun Wang
- Henan Provincial People's Hospital & Zhengzhou University People's Hospital, Zhengzhou 450003, P. R. China
- Henan Key Laboratory of Neurological Imaging, Zhengzhou University, Zhengzhou 450003, P. R. China
| |
Collapse
|
7
|
Recent Trends of microRNA Significance in Pediatric Population Glioblastoma and Current Knowledge of Micro RNA Function in Glioblastoma Multiforme. Int J Mol Sci 2020; 21:ijms21093046. [PMID: 32349263 PMCID: PMC7246719 DOI: 10.3390/ijms21093046] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system tumors are a significant problem for modern medicine because of their location. The explanation of the importance of microRNA (miRNA) in the development of cancerous changes plays an important role in this respect. The first papers describing the presence of miRNA were published in the 1990s. The role of miRNA has been pointed out in many medical conditions such as kidney disease, diabetes, neurodegenerative disorder, arthritis and cancer. There are several miRNAs responsible for invasiveness, apoptosis, resistance to treatment, angiogenesis, proliferation and immunology, and many others. The research conducted in recent years analyzing this group of tumors has shown the important role of miRNA in the course of gliomagenesis. These particles seem to participate in many stages of the development of cancer processes, such as proliferation, angiogenesis, regulation of apoptosis or cell resistance to cytostatics.
Collapse
|
8
|
Siedler DG, Beechey JC, Jessup PJ, Thani NB. Infantile Optic Pathway Glioblastoma. World Neurosurg 2019; 129:172-175. [PMID: 31158532 DOI: 10.1016/j.wneu.2019.05.236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/24/2019] [Accepted: 05/25/2019] [Indexed: 11/16/2022]
Abstract
BACKGROUND Optic pathway gliomas and glioblastomas remain a rare entity within the infant population. CASE DESCRIPTION We outline the case of a 6-month-old female who presented with failure to thrive, nystagmus and features of raised intracranial pressure. Subsequent magnetic resonance imaging demonstrated an infiltrating tumor radiating from the optic nerves bilaterally. She underwent emergent ventriculoperitoneal shunting and biopsy. Histology confirmed a World Health Organization grade IV glioblastoma. CONCLUSIONS The patient remained clinically and radiologically stable at 1 year. Optic pathway glioblastoma in this population is a previously undescribed entity that requires multidisciplinary input to guide ongoing therapy.
Collapse
Affiliation(s)
- Declan G Siedler
- Department of Neurosurgery, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Jessica C Beechey
- Department of Anatomical Pathology, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Peter J Jessup
- Department of Anatomical Pathology, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Nova B Thani
- Department of Neurosurgery, Royal Hobart Hospital, Hobart, Tasmania, Australia.
| |
Collapse
|
9
|
Fangusaro J, Onar-Thomas A, Young Poussaint T, Wu S, Ligon AH, Lindeman N, Banerjee A, Packer RJ, Kilburn LB, Goldman S, Pollack IF, Qaddoumi I, Jakacki RI, Fisher PG, Dhall G, Baxter P, Kreissman SG, Stewart CF, Jones DTW, Pfister SM, Vezina G, Stern JS, Panigrahy A, Patay Z, Tamrazi B, Jones JY, Haque SS, Enterline DS, Cha S, Fisher MJ, Doyle LA, Smith M, Dunkel IJ, Fouladi M. Selumetinib in paediatric patients with BRAF-aberrant or neurofibromatosis type 1-associated recurrent, refractory, or progressive low-grade glioma: a multicentre, phase 2 trial. Lancet Oncol 2019; 20:1011-1022. [PMID: 31151904 DOI: 10.1016/s1470-2045(19)30277-3] [Citation(s) in RCA: 336] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/16/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Paediatric low-grade glioma is the most common CNS tumour of childhood. Although overall survival is good, disease often recurs. No single universally accepted treatment exists for these patients; however, standard cytotoxic chemotherapies are generally used. We aimed to assess the activity of selumetinib, a MEK1/2 inhibitor, in these patients. METHODS The Pediatric Brain Tumor Consortium performed a multicentre, phase 2 study in patients with paediatric low-grade glioma in 11 hospitals in the USA. Patients aged 3-21 years with a Lansky or Karnofsky performance score greater than 60 and the presence of recurrent, refractory, or progressive paediatric low-grade glioma after at least one standard therapy were eligible for inclusion. Patients were assigned to six unique strata according to histology, tumour location, NF1 status, and BRAF aberration status; herein, we report the results of strata 1 and 3. Stratum 1 comprised patients with WHO grade I pilocytic astrocytoma harbouring either one of the two most common BRAF aberrations (KIAA1549-BRAF fusion or the BRAFV600E [Val600Glu] mutation). Stratum 3 comprised patients with any neurofibromatosis type 1 (NF1)-associated paediatric low-grade glioma (WHO grades I and II). Selumetinib was provided as capsules given orally at the recommended phase 2 dose of 25 mg/m2 twice daily in 28-day courses for up to 26 courses. The primary endpoint was the proportion of patients with a stratum-specific objective response (partial response or complete response), as assessed by the local site and sustained for at least 8 weeks. All responses were reviewed centrally. All eligible patients who initiated treatment were evaluable for the activity and toxicity analyses. Although the trial is ongoing in other strata, enrolment and planned follow-up is complete for strata 1 and 3. This trial is registered with ClinicalTrials.gov, number NCT01089101. FINDINGS Between July 25, 2013, and June 12, 2015, 25 eligible and evaluable patients were accrued to stratum 1, and between Aug 28, 2013, and June 25, 2015, 25 eligible and evaluable patients were accrued to stratum 3. In stratum 1, nine (36% [95% CI 18-57]) of 25 patients achieved a sustained partial response. The median follow-up for the 11 patients who had not had a progression event by Aug 9, 2018, was 36·40 months (IQR 21·72-45·59). In stratum 3, ten (40% [21-61]) of 25 patients achieved a sustained partial response; median follow-up was 48·60 months (IQR 39·14-51·31) for the 17 patients without a progression event by Aug 9, 2018. The most frequent grade 3 or worse adverse events were elevated creatine phosphokinase (five [10%]) and maculopapular rash (five [10%]). No treatment-realted deaths were reported. INTERPRETATION Selumetinib is active in recurrent, refractory, or progressive pilocytic astrocytoma harbouring common BRAF aberrations and NF1-associated paediatric low-grade glioma. These results show that selumetinib could be an alternative to standard chemotherapy for these subgroups of patients, and have directly led to the development of two Children's Oncology Group phase 3 studies comparing standard chemotherapy to selumetinib in patients with newly diagnosed paediatric low-grade glioma both with and without NF1. FUNDING National Cancer Institute Cancer Therapy Evaluation Program, the American Lebanese Syrian Associated Charities, and AstraZeneca.
Collapse
Affiliation(s)
- Jason Fangusaro
- Department of Hematology, Oncology, and Stem Cell Transplantation, Children's Healthcare of Atlanta and Emory University, Atlanta, GA, USA.
| | - Arzu Onar-Thomas
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Shengjie Wu
- Department of Biostatistics, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Azra H Ligon
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Neal Lindeman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anuradha Banerjee
- Center for Cancer and Blood Disorders, University of California, San Francisco, San Francisco, CA, USA
| | - Roger J Packer
- Department of Neurology, Children's National Medical Center, Washington, DC, USA
| | - Lindsay B Kilburn
- Department of Haematology and Oncology, Children's National Medical Center, Washington, DC, USA
| | - Stewart Goldman
- Department of Haematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Ian F Pollack
- Department of Neurosurgery, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Ibrahim Qaddoumi
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Regina I Jakacki
- Department of Hematology and Oncology, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Paul G Fisher
- Department of Neurology, Stanford University Medical Center, Palo Alto, CA, USA
| | - Girish Dhall
- Department of Hematology and Oncology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Patricia Baxter
- Department of Hematology and Oncology, Texas Children's Hospital, Houston, TX, USA
| | - Susan G Kreissman
- Department of Hematology and Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Clinton F Stewart
- Department of Pharmaceutical Science, St Jude Children's Research Hospital, Memphis, TN, USA
| | - David T W Jones
- Department of Pediatric Glioma Research Group, Hopp Children's Cancer Center Heidelberg (KiTZ) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan M Pfister
- Department of Pediatric Neuro-Oncology, Hopp Children's Cancer Center Heidelberg (KiTZ) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gilbert Vezina
- Department of Radiology, Children's National Medical Center, Washington, DC, USA
| | - Jessica S Stern
- Department of Radiology, Ann and Robert H Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Ashok Panigrahy
- Department of Radiology, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zoltan Patay
- Department of Diagnostic Imaging, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Benita Tamrazi
- Department of Radiology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Jeremy Y Jones
- Department of Radiology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Sofia S Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David S Enterline
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Soonmee Cha
- Department of Radiology, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J Fisher
- Department of Pediatric Oncology, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Laurence Austin Doyle
- Investigational Drug Branch, National Cancer Institute and Cancer Therapy Evaluation Program, Bethesda, MD, USA
| | - Malcolm Smith
- Clinical Investigation Branch, National Cancer Institute and Cancer Therapy Evaluation Program, Bethesda, MD, USA
| | - Ira J Dunkel
- Department of Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Maryam Fouladi
- Department of Haematology and Oncology, Cincinnati Children's Hospital, Cincinnati, OH, USA
| |
Collapse
|
10
|
Delays in Diagnosis of Pediatric Histologically Confirmed Sellar Germ Cell Tumors in China: A Retrospective Risk Factor Analysis. World Neurosurg 2019; 122:e472-e479. [DOI: 10.1016/j.wneu.2018.10.082] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 02/07/2023]
|
11
|
Chemical modulation of autophagy as an adjunct to chemotherapy in childhood and adolescent brain tumors. Oncotarget 2018; 9:35266-35277. [PMID: 30443293 PMCID: PMC6219655 DOI: 10.18632/oncotarget.26186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 08/27/2018] [Indexed: 02/07/2023] Open
Abstract
Brain tumors are the leading cause of cancer-related death in children and are the most challenging childhood cancer in relation to diagnosis, treatment, and outcome. One potential novel strategy to improve outcomes in cancer involves the manipulation of autophagy, a fundamental process in all cells. In cancer, autophagy can be thought of as having a "Janus"-like duality. On one face, especially in the early phases of cancer formation, autophagy can act as a cellular housekeeper to eliminate damaged organelles and recycle macromolecules, thus acting as tumor suppressor. On the other face, at later stages of tumor progression, autophagy can function as a pro-survival pathway in response to metabolic stresses such as nutrient depravation, hypoxia and indeed to chemotherapy itself, and can support cell growth by supplying much needed energy. In the context of chemotherapy, autophagy may, in some cases, mediate resistance to treatment. We present an overview of the relevance of autophagy in central nervous system tumors including how its chemical modulation can serve as a useful adjunct to chemotherapy, and use this knowledge to consider how targeting of autophagy may be relevant in pediatric brain tumors.
Collapse
|
12
|
Lassaletta A, Strother D, Laperriere N, Hukin J, Vanan MI, Goddard K, Lafay-Cousin L, Johnston DL, Zelcer S, Zapotocky M, Rajagopal R, Ramaswamy V, Hawkins C, Tabori U, Huang A, Bartels U, Bouffet E. Reirradiation in patients with diffuse intrinsic pontine gliomas: The Canadian experience. Pediatr Blood Cancer 2018; 65:e26988. [PMID: 29369515 DOI: 10.1002/pbc.26988] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Clinical trials have failed to demonstrate a survival benefit of adjuvant chemotherapy in diffuse intrinsic pontine gliomas (DIPG). Radiation therapy (RT) is the only effective treatment thus far and reirradiation (rRT) has become an option at the time of progression. The aim of this study was to review the Canadian experience of DIPG rRT with a focus on the safety and possible efficacy of this approach. METHOD We retrospectively reviewed the demographic, clinical, and RT data of patients with DIPG treated in Canada with rRT. RESULTS Since January 2011, we identified 16 patients with progressive DIPG who received rRT. Median time from diagnosis to progression was 10.5 months (range, 4-37 months). rRT was given focally in 14 patients at a dose ranging from 21.6 to 36 Gy. rRT was well tolerated by all children but one. All but three patients showed neurological improvement. With a median follow-up from original diagnosis of 19.2 months, all patients died, with a median time from rRT to death of 6.48 months (range, 3.83-13.26 months). When compared to a historic cohort of 46 consecutive patients, the median time from progression to death was 92 days in the non-reirradiated patients versus 218 days in the reirradiated ones (P = 0.0001). CONCLUSION In this limited experience, rRT was safe and feasible in patients with progressive DIPG, providing neurological improvement and a prolonged life span in most patients. Prospective Canadian rRT protocols are ongoing to further assess the benefit of this approach, including quality of life assessment.
Collapse
Affiliation(s)
| | - Douglas Strother
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Juliette Hukin
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | | | - Karen Goddard
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Lucie Lafay-Cousin
- Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Shayna Zelcer
- Children's Hospital of Western Ontario, London, Ontario, Canada
| | | | | | | | | | - Uri Tabori
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annie Huang
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ute Bartels
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Eric Bouffet
- The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Zhang Y, Zhu H, Deng K, Ma W, Wang Y, Sun J, Lian X, Pan H, Wang R, Yao Y. Results of Biopsy-Proven Sellar Germ Cell Tumors: Nine Years' Experience in a Single Center. World Neurosurg 2018; 112:e229-e239. [DOI: 10.1016/j.wneu.2018.01.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/01/2018] [Accepted: 01/04/2018] [Indexed: 01/28/2023]
|
14
|
Mascelli S, Nozza P, Jones DTW, Colin C, Pistorio A, Milanaccio C, Ravegnani M, Consales A, Witt O, Morana G, Cama A, Capra V, Biassoni R, Pfister SM, Figarella-Branger D, Garrè ML, Raso A. TP53 codon 72 polymorphism may predict early tumour progression in paediatric pilocytic astrocytoma. Oncotarget 2018; 7:47918-47926. [PMID: 27374106 PMCID: PMC5216988 DOI: 10.18632/oncotarget.10295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/10/2016] [Indexed: 12/13/2022] Open
Abstract
Pilocytic astrocytoma and ganglioglioma may occur in inaccessible or surgically difficult areas. In case of incomplete resection, the availability of biological predictors of tumour progression could be particularly important. To this end, an analysis of p53 codon 72 polymorphism and assessment of its role as prognostic marker were performed. The status of the p53 Arg72Pro polymorphism was evaluated by pyrosequencing method in a multicenter cohort of 170 paediatric patients. Genotype/phenotype associations were investigated either by means of bivariate or multivariate analyses. In the partially resected pilocytic astrocytomas, the Arg/Arg variant predicts early tumour progression (median survival time: 23.1 months) and is associated with poor event-free survival (p value = 0.0009). This finding remains true also in case of adjuvant therapies, with a 5-year event-free survival of 30.6% for cases with Arg/Arg variant vs. 78.7% for those with other genotypes. There is no association between ganglioglioma and the polymorphism. The assessment of Arg/Arg variant could improve the management of pilocytic astrocytoma. TP53 codon 72 analysis could distinguish low-risk cases, in which surgery could be conservative, from high-risk cases needing an aggressive surgery plan.
Collapse
Affiliation(s)
- Samantha Mascelli
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Paolo Nozza
- UOC Anatomia Patologica, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - David T W Jones
- Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Carole Colin
- CRO2 UMR_S911, Inserm, Aix-Marseille Université, 13385 Marseille, France
| | - Angela Pistorio
- Epidemiologia, Biostatistica e Comitati, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Claudia Milanaccio
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Marcello Ravegnani
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Alessandro Consales
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Olaf Witt
- Department of Paediatric Oncology, Haematology and Immunology, University of Heidelberg, Heidelberg, 69120 Heidelberg, Germany
| | - Giovanni Morana
- Neuroradiologia Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Armando Cama
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Valeria Capra
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Roberto Biassoni
- Laboratory Molecular Medicine, Translational Medicine Department, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Stefan M Pfister
- Division of Paediatric Neurooncology, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany.,Department of Paediatric Oncology, Haematology and Immunology, University of Heidelberg, Heidelberg, 69120 Heidelberg, Germany
| | - Dominique Figarella-Branger
- CRO2 UMR_S911, Inserm, Aix-Marseille Université, 13385 Marseille, France.,APHM, Hôpital de la Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13385 Marseille, France
| | - Maria Luisa Garrè
- Centro di Neuro-Oncologia, Istituto Giannina Gaslini, 16147 Genoa, Italy
| | - Alessandro Raso
- Dipartimento Testa, Collo e Neuroscienze, Istituto Giannina Gaslini, 16147 Genoa, Italy
| |
Collapse
|
15
|
Liang ML, Hsieh TH, Ng KH, Tsai YN, Tsai CF, Chao ME, Liu DJ, Chu SS, Chen W, Liu YR, Liu RS, Lin SC, Ho DMT, Wong TT, Yang MH, Wang HW. Downregulation of miR-137 and miR-6500-3p promotes cell proliferation in pediatric high-grade gliomas. Oncotarget 2017; 7:19723-37. [PMID: 26933822 PMCID: PMC4991414 DOI: 10.18632/oncotarget.7736] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/18/2016] [Indexed: 01/22/2023] Open
Abstract
Pediatric high-grade gliomas (pHGGs) are aggressive brain tumors affecting children, and outcomes have remained dismal, even with access to new multimodal therapies. In this study, we compared the miRNomes and transcriptomes of pediatric low- (pLGGs) and high-grade gliomas (pHGGs) using small RNA sequencing (smRNA-Seq) and gene expression microarray, respectively. Through integrated bioinformatics analyses and experimental validation, we identified miR-137 and miR-6500-3p as significantly downregulated in pHGGs. miR-137 or miR-6500-3p overexpression reduced cell proliferation in two pHGG cell lines, SF188 and UW479. CENPE, KIF14 and NCAPG levels were significantly higher in pHGGs than pLGGs, and were direct targets of miR-137 or miR-6500-3p. Furthermore, knockdown of CENPE, KIF14 or NCAPG combined with temozolomide treatment resulted in a combined suppressive effect on pHGG cell proliferation. In summary, our results identify novel mRNA/miRNA interactions that contribute to pediatric glioma malignancy and represent potential targets for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Muh-Lii Liang
- Institutes of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tsung-Han Hsieh
- PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Comprehensive Cancer Center of Taipei Medical University, Taipei Medical University, Taipei, Taiwan
| | - Kim-Hai Ng
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Ya-Ni Tsai
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Fong Tsai
- Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan
| | - Meng-En Chao
- Institutes of Clinical Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Da-Jung Liu
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shing-Shiung Chu
- Institutes of Clinical Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wan Chen
- Institutes of Clinical Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Comprehensive Cancer Center of Taipei Medical University, Taipei Medical University, Taipei, Taiwan.,Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Ren-Shyan Liu
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,National PET/Cyclotron Center, Department of Nuclear Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Molecular and Genetic Imaging Core/Taiwan Mouse Clinic National Comprehensive Mouse Phenotyping and Drug Testing Center, Taipei, Taiwan
| | - Shih-Chieh Lin
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Donald Ming-Tak Ho
- School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tai-Tong Wong
- Division of Pediatric Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan.,Institutes of Clinical Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurosurgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institutes of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Cancer Research Center & Genome Research Center, National Yang-Ming University, Taipei, Taiwan.,Division of Hematology-Oncology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Immunity and Inflammation Research Center, National Yang-Ming University, Taipei, Taiwan.,Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Hsei-Wei Wang
- Institutes of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan.,Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan.,Institute of Biomedical Informatics, National Yang-Ming University, Taipei, Taiwan.,Cancer Research Center & Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
16
|
Diwanji TP, Engelman A, Snider JW, Mohindra P. Epidemiology, diagnosis, and optimal management of glioma in adolescents and young adults. ADOLESCENT HEALTH MEDICINE AND THERAPEUTICS 2017; 8:99-113. [PMID: 28989289 PMCID: PMC5624597 DOI: 10.2147/ahmt.s53391] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neoplasms of the central nervous system (CNS) are the most frequently encountered solid tumors of childhood, but are less common in adolescents and young adults (AYA), aged 15–39 years. Gliomas account for 29%–35% of the CNS tumors in AYA, with approximately two-thirds being low-grade glioma (LGG) and the remaining being high-grade glioma (HGG). We review the epidemiology, work-up, and management of LGG and HGG, focusing on the particular issues faced by the AYA population relative to pediatric and adult populations. Visual pathway glioma and brainstem glioma, which represent unique clinical entities, are only briefly discussed. As a general management approach for both LGG and HGG, maximal safe resection should be attempted. AYA with LGG who undergo gross total resection (GTR) may be safely observed. As age increases and the risk factors for recurrence accumulate, adjuvant therapy should be more strongly considered with a strong consideration of advanced radiation techniques such as proton beam therapy to reduce long-term radiation-related toxicity. Recent results also suggest survival advantage for adult patients with the use of adjuvant chemotherapy when radiation is indicated. Whenever possible, AYA patients with HGG should be enrolled in a clinical trial for the benefit of centralized genetic and molecular prognostic review and best clinical care. Chemoradiation should be offered to all World Health Organization grade IV patients with concurrent and adjuvant chemotherapy after maximal safe resection. Younger adolescents with GTR of grade III lesions may consider radiotherapy alone or sequential radiotherapy and chemotherapy if unable to tolerate concurrent treatment. A more comprehensive classification of gliomas integrating pathology and molecular data is emerging, and this integrative strategy offers the potential to be more accurate and reproducible in guiding diagnostic, prognostic, and management decisions.
Collapse
Affiliation(s)
- Tejan P Diwanji
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alexander Engelman
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James W Snider
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pranshu Mohindra
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Klonou A, Piperi C, Gargalionis AN, Papavassiliou AG. Molecular Basis of Pediatric Brain Tumors. Neuromolecular Med 2017; 19:256-270. [DOI: 10.1007/s12017-017-8455-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 07/21/2017] [Indexed: 01/03/2023]
|
18
|
Abstract
Astrocytomas (gliomas) are the most common primary brain tumors among adults and second most frequent neoplasm among children. New ideas and novel approaches are being explored world over with aim to devise better management strategeies for this deadly pathological state. We searched the electronic database PubMed for pre-clinical as well as clinical controlled trials reporting importance of various therapeutic drugs against gliomas. It was observed clearly that this approach of using therapeutic drugs is clearly evolving and has been observed to be promising future therapeutic avenue against gliomas. The searched literature on whole revealed that although gliomas are treated aggressively with surgery, chemotherapy and radiation, treatment resistance, drug toxicity and poor response rates among pediatric glioma patients, continue to drive the need to discover new and more effective chemotherapeutic agents. The present review is focused on the latest updates in therapeutic drugs against gliomas in pediatric patients. The important chemo-therapeutics discussed in this review included alkylating agents like temoxolomide, derivatives of platinum, nitrosoureas, topoisomerases, angiogenesis inhibitors and cytomegalovirus as therapeutic agents.
Collapse
|
19
|
Garcia MA, Solomon DA, Haas-Kogan DA. Exploiting molecular biology for diagnosis and targeted management of pediatric low-grade gliomas. Future Oncol 2016; 12:1493-506. [PMID: 27072750 PMCID: PMC4915741 DOI: 10.2217/fon-2016-0039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/29/2016] [Indexed: 12/14/2022] Open
Abstract
The majority of brain tumors arising in children are low-grade gliomas. Although historically categorized together as pediatric low-grade gliomas (PLGGs), there is significant histologic and genetic diversity within this group. In general, prognosis for PLGGs is excellent, and limitation of sequelae from tumor and treatment is paramount. Advances in high-throughput genetic sequencing and gene expression profiling are fundamentally changing the way PLGGs are classified and managed. Here, we review the histologic subtypes and highlight how recent advances in elucidating the molecular pathogenesis of these tumors have refined diagnosis and prognostication. Additionally, we discuss how characterizing specific genetic alterations has paved the way for the rational use of targeted therapies that are currently in various phase clinical trials.
Collapse
Affiliation(s)
- Michael A Garcia
- Department of Radiation Oncology, University of California, San Francisco, CA, USA
| | - David A Solomon
- Division of Neuropathology, Department of Pathology, University of California, San Francisco, CA, USA
| | - Daphne A Haas-Kogan
- Department of Radiation Oncology, Harvard Medical School, Dana-Farber Cancer Institute, Brigham & Women's Hospital, Boston Children's Hospital, MA, USA
| |
Collapse
|
20
|
Peron A, Vignoli A, La Briola F, Volpi A, Montanari E, Morenghi E, Ghelma F, Bulfamante G, Cefalo G, Canevini MP. Do patients with tuberous sclerosis complex have an increased risk for malignancies? Am J Med Genet A 2016; 170:1538-44. [PMID: 27061015 DOI: 10.1002/ajmg.a.37644] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/20/2016] [Indexed: 02/01/2023]
Abstract
Tuberous Sclerosis Complex (TSC) is generally characterized by the presence of benign tumors, but some patients with malignancies have been reported in the literature. We examined a large Italian TSC population (240 individuals followed from 2001 to 2015, aged 3 months-74 years), assessing the frequency of malignancies to determine whether there is an increased risk for cancer in this disorder, and looking for possible features associated with the development of neoplasia. Fifteen patients had malignancies (6.25%); median age at diagnosis was 37.5 years (range of 1.6-58). Five of seven renal tumors were renal cell carcinomas. Eight patients had a non-renal malignancy (3.3%), but we did not find a more prevalent type of cancer. No patient developed more than one malignancy. The prevalence of all malignant tumors was compatible with the prevalence in the general population (5.6%, 95%CI 2.99-9.31%, vs. 4.4% in Italy). Median age at cancer diagnosis was lower (37.5 years, 95%CI 28.6-44.7, vs. 66.0 years). Two patients (13.3%) died of their cancer, while outcome was favorable in the remaining individuals. Malignant tumors were more frequently diagnosed in patients with mutations in TSC1 when compared to TSC2 and patients with no mutation identified (P = 0.032). Our study demonstrated that TSC patients do not seem to have an increased risk for malignancies besides renal cell carcinoma. However, when cancer develops, age at diagnosis is lower than in the general population, and malignant tumors are more frequently diagnosed in patients with mutations in TSC1. Further studies are needed to confirm these data. ©2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Angela Peron
- Child Neurology Unit-Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Aglaia Vignoli
- Child Neurology Unit-Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca La Briola
- Child Neurology Unit-Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Angela Volpi
- Nephrology Unit, San Paolo Hospital, Milan, Italy
| | - Emanuele Montanari
- Urology Unit, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Emanuela Morenghi
- Biostatististics Unit, Istituto Clinico Humanitas, Rozzano, Milan, Italy
| | - Filippo Ghelma
- DAMA Unit (Disabled Advanced Medical Assistance), San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Gaetano Bulfamante
- Human Pathology Unit, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Graziella Cefalo
- Pediatrics Department, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria Paola Canevini
- Child Neurology Unit-Epilepsy Center, San Paolo Hospital, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
21
|
Abstract
INTRODUCTION The purpose of this review is to document the various types of astrocytoma that occur in the fetus and neonate, their locations, initial findings, pathology, and outcome. Data are presented that show which patients are likely to survive or benefit from treatment compared with those who are unlikely to respond. MATERIALS AND METHODS One hundred one fetal and neonatal tumors were collected from the literature for study. RESULTS Macrocephaly and an intracranial mass were the most common initial findings. Overall, hydrocephalus and intracranial hemorrhage were next. Glioblastoma (GBM) was the most common neoplasm followed in order by subependymal giant cell astrocytoma (SEGA), low-grade astrocytoma, anaplastic astrocytoma, and desmoplastic infantile astrocytoma (DIA). Tumors were detected most often toward the end of the third trimester of pregnancy. CONCLUSION A number of patients were considered inoperable since their tumor occupied much of the intracranial cavity involving large areas of the brain. High-grade astrocytomas were more common than low-grade ones in this review. Fetuses and neonates with astrocytoma have a mixed prognosis ranging from as low as 20 % (GBM) to a high of 90 %. The overall survival was 47/101 or 46 %.
Collapse
|
22
|
McAbee JH, Modica J, Thompson CJ, Broniscer A, Orr B, Choudhri AF, Boop FA, Klimo P. Cervicomedullary tumors in children. J Neurosurg Pediatr 2015; 16:357-66. [PMID: 26114990 DOI: 10.3171/2015.5.peds14638] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Cervicomedullary tumors (CMTs) represent a heterogeneous group of intrinsic neoplasms that are typically low grade and generally carry a good prognosis. This single-institution study was undertaken to document the outcomes and current treatment philosophy for these challenging neoplasms. METHODS The charts of all pediatric patients with CMTs who received treatment at St. Jude Children's Research Hospital between January 1988 and May 2013 were retrospectively reviewed. Demographic, surgical, clinical, radiological, pathological, and survival data were collected. Treatment-free survival and overall survival were estimated, and predictors of recurrence were analyzed. RESULTS Thirty-one children (16 boys, 15 girls) with at least 12 months of follow-up data were identified. The median age at diagnosis was 6 years (range 7 months-17 years) and the median follow-up was 4.3 years. Low-grade tumors (Grade I or II) were present in 26 (84%) patients. Thirty patients underwent either a biopsy alone or resection, with the majority of patients undergoing biopsy only (n = 12, 39%) or subtotal resection (n = 14, 45%). Only 4 patients were treated solely with resection; 21 patients received radiotherapy alone or in combination with other treatments. Recurrent tumor developed in 14 children (45%) and 4 died as a result of their malignancy. A high-grade pathological type was the only independent variable that predicted recurrence. The 5- and 10-year treatment-free survival estimates are 64.7% and 45.3%, respectively. The 5- and 10-year overall survival estimate is 86.7%. CONCLUSIONS Children with CMTs typically have low-grade neoplasms and consequently long-term survival, but high risk of recurrence. Therapy should be directed at achieving local tumor control while preserving and even restoring neurological function.
Collapse
Affiliation(s)
- Joseph H McAbee
- School of Medicine, Wake Forest University, Winston-Salem, North Carolina
| | - Joseph Modica
- University at Buffalo School of Medicine and Biomedical Sciences, Buffalo, New York
| | - Clinton J Thompson
- George Washington University Milken Institute School of Public Health, Washington, DC; and
| | - Alberto Broniscer
- Department of Oncology, St. Jude Children's Research Hospital;,Department of Pediatrics, University of Tennessee Health Science Center
| | - Brent Orr
- Department of Pathology, St. Jude Children's Research Hospital
| | - Asim F Choudhri
- Department of Neurosurgery, University of Tennessee Health Science Center;,Department of Radiology, University of Tennessee Health Science Center;,Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital
| | - Frederick A Boop
- Department of Neurosurgery, University of Tennessee Health Science Center;,Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital;,Department of Surgery, St. Jude Children's Research Hospital; and.,Semmes-Murphey Neurologic & Spine Institute, Memphis, Tennessee
| | - Paul Klimo
- Department of Neurosurgery, University of Tennessee Health Science Center;,Le Bonheur Neuroscience Institute, Le Bonheur Children's Hospital;,Department of Surgery, St. Jude Children's Research Hospital; and.,Semmes-Murphey Neurologic & Spine Institute, Memphis, Tennessee
| |
Collapse
|
23
|
Vignoli A, Lesma E, Alfano RM, Peron A, Scornavacca GF, Massimino M, Schiavello E, Ancona S, Cerati M, Bulfamante G, Gorio A, Canevini MP. Glioblastoma multiforme in a child with tuberous sclerosis complex. Am J Med Genet A 2015; 167A:2388-93. [PMID: 25946256 DOI: 10.1002/ajmg.a.37158] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/27/2015] [Indexed: 12/27/2022]
Abstract
Tuberous Sclerosis Complex (TSC) is characterized by the presence of benign tumors in the brain, kidneys, heart, eyes, lungs, and skin. The typical brain lesions are cortical tubers, subependimal nodules and subependymal giant-cell astrocytomas. The occurrence of malignant astrocytomas such as glioblastoma is rare. We report on a child with a clinical diagnosis of TSC and a rapidly evolving glioblastoma multiforme. Genetic analysis identified a de novo mutation in TSC2. Molecular characterization of the tumor was performed and discussed, as well as a review of the literature where cases of TSC and glioblastoma multiforme are described. Although the co-occurrence of TSC and glioblastoma multiforme seems to be rare, this possible association should be kept in mind, and proper clinical and radiological follow up should be recommended in these patients.
Collapse
Affiliation(s)
- Aglaia Vignoli
- Child Neurology Unit - Epilepsy Center, Department of Health Science, University of Milan, San Paolo Hospital, Milano, Italy
| | - Elena Lesma
- Laboratories of Pharmacology, Department of Health Science, University of Milan, Milano, Italy
| | - Rosa Maria Alfano
- Department of Human Pathology, Cytogenetic and Molecular Pathology, Department of Health Science, San Paolo Hospital, Milano, Italy
| | - Angela Peron
- Child Neurology Unit - Epilepsy Center, Department of Health Science, University of Milan, San Paolo Hospital, Milano, Italy
| | - Giulia Federica Scornavacca
- Child Neurology Unit - Epilepsy Center, Department of Health Science, University of Milan, San Paolo Hospital, Milano, Italy
| | - Maura Massimino
- Pediatric Unit, Department of Hematology and Pediatric Oncoematology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milano, Italy
| | - Elisabetta Schiavello
- Pediatric Unit, Department of Hematology and Pediatric Oncoematology, Fondazione IRCCS, Istituto Nazionale dei Tumori, Milano, Italy
| | - Silvia Ancona
- Laboratories of Pharmacology, Department of Health Science, University of Milan, Milano, Italy
| | - Michele Cerati
- Department of Pathology, Ospedale di Circolo, Varese, Italy
| | - Gaetano Bulfamante
- Department of Human Pathology, Cytogenetic and Molecular Pathology, Department of Health Science, San Paolo Hospital, Milano, Italy
| | - Alfredo Gorio
- Laboratories of Pharmacology, Department of Health Science, University of Milan, Milano, Italy
| | - Maria Paola Canevini
- Child Neurology Unit - Epilepsy Center, Department of Health Science, University of Milan, San Paolo Hospital, Milano, Italy
| |
Collapse
|
24
|
Torres-Martín M, Peña-Granero C, Carceller F, Gutiérrez M, Burbano RR, Pinto GR, Castresana JS, Melendez B, Rey JA. Homozygous deletion of TNFRSF4, TP73, PPAP2B and DPYD at 1p and PDCD5 at 19q identified by multiplex ligation-dependent probe amplification (MLPA) analysis in pediatric anaplastic glioma with questionable oligodendroglial component. Mol Cytogenet 2014; 7:1. [PMID: 24387276 PMCID: PMC3905963 DOI: 10.1186/1755-8166-7-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 12/11/2013] [Indexed: 02/03/2023] Open
Abstract
Background Pediatric oligodendrogliomas are rare and appear to show a different molecular profile from adult tumors. Some gliomas display allelic losses at 1p/19q in pediatric patients, although less frequently than in adult patients, but this is rare in tumors with an oligodendroglial component. The molecular basis of this genomic abnormality is unknown in pediatric gliomas, but it represents a relatively common finding in pediatric oligodendroglioma-like neoplasms with leptomeningeal dissemination. Results Multiplex ligation-dependent probe amplification (MLPA) analysis using SALSA P088-B1 for the analysis of the 1p/19q allelic constitution in a pediatric anaplastic (oligodendro)-glioma showed homozygous co-deletion for markers: TNFRSF4 (located at 1p36.33), TP73 (1p36.32), PPAP2B (1pter-p22.1), DPYD (1p21.3), and PDCD5 (19q13.12), and hemizygous deletion of BAX (19q13.3-q13.4). No sequence changes for R132 and R172 of the IDH1/2 genes were identified. Conclusions The molecular findings in this pediatric anaplastic glioma do not allow for a clearly definitive pathological diagnosis. However, the findings provide data on a number of 1p/19q genomic regions that, because of homozygotic deletion, might be the location of genes that are important for the development and clinical evolution of some malignant gliomas in children.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan A Rey
- Molecular Neuro-oncogenetics Laboratory, Research Unit-Unidad de Investigación, Hospital Universitario La Paz, Paseo de la Castellana 261, 28046 Madrid, Spain.
| |
Collapse
|
25
|
Wozel G, Blasum C. Dapsone in dermatology and beyond. Arch Dermatol Res 2013; 306:103-24. [PMID: 24310318 PMCID: PMC3927068 DOI: 10.1007/s00403-013-1409-7] [Citation(s) in RCA: 215] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/30/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022]
Abstract
Dapsone (4,4′-diaminodiphenylsulfone) is an aniline derivative belonging to the group of synthetic sulfones. In 1937 against the background of sulfonamide era the microbial activity of dapsone has been discovered. Shortly thereafter, the use of dapsone to treat non-pathogen-caused diseases revealed alternate antiinflammatory mechanisms that initially were elucidated by inflammatory animal models. Thus, dapsone clearly has dual functions of both: antimicrobial/antiprotozoal effects and anti-inflammatory features similarly to non-steroidal anti-inflammatory drugs. The latter capabilities primarily were used in treating chronic inflammatory disorders. Dapsone has been investigated predominantly by in vitro methods aiming to get more insights into the effect of dapsone to inflammatory effector cells, cytokines, and/or mediators, such as cellular toxic oxygen metabolism, myoloperoxidase-/halogenid system, adhesion molecules, chemotaxis, membrane-associated phospholipids, prostaglandins, leukotrienes, interleukin-8, tumor necrosis factor α, lymphocyte functions, and tumor growth. Moreover, attention has been paid to mechanisms by which dapsone mediates effects in more complex settings like impact of lifespan, stroke, glioblastoma, or as anticonvulsive agent. Additionally, there are some dermatological investigations in human being using dapsone and its metabolites (e.g., leukotriene B4-induced chemotaxis, ultraviolet-induced erythema). It could be established that dapsone metabolites by their own have anti-inflammatory properties. Pharmacology and mechanisms of action are determining factors for clinical use of dapsone chiefly in neutrophilic and/or eosinophilic dermatoses and in chronic disorders outside the field of dermatology. The steroid-sparing effect of dapsone is useful for numerous clinical entities. Future avenues of investigations will provide more information on this fascinating and essential agent.
Collapse
Affiliation(s)
- Gottfried Wozel
- Study Centre for Clinical Trials, Dermatology, Gesellschaft für Wissens- und Technologietransfer der Technischen Universität Dresden mbH, Blasewitzer Str. 43, 01307 Dresden, Germany
| | - Christian Blasum
- Private Practice of Dermatology, Marktplatz 25, 73728 Esslingen, Germany
| |
Collapse
|
26
|
Ajeawung NF, Faure R, Jones C, Kamnasaran D. Preclinical evaluation of dipotassium bisperoxo (picolinato) oxovanadate V for the treatment of pediatric low-grade gliomas. Future Oncol 2013; 9:1215-29. [PMID: 23902250 DOI: 10.2217/fon.13.73] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIM The treatment of pediatric low-grade gliomas with current treatment modalities still remains ineffective among a subset of patients; hence, justifying the need to further investigate more effective therapies. Dipotassium bisperoxo (picolinato) oxovanadate V (Bpv[pic]), is a derivative of the trace metal vanadium and a potent inhibitor of protein tyrosine phosphatases, which are important mediators of oncogenic and tumor suppressive activities in cancers. In this study, we undertook a preclinical evaluation of the antineoplastic functions of Bpv(pic) in the treatment of pediatric low-grade gliomas. MATERIALS & METHODS We utilized pediatric low-grade glioma cell lines (Res186, Res259 and R286) in a wide variety of cancer assays to determine whether Bpv(pic) can abrogate the neoplastic properties of these cells. RESULTS Our preclinical evaluation of the antineoplastic properties of Bpv(pic) in pediatric low-grade gliomas reveals a significant dose-dependent decrease in cell viability as a consequence of decreased proliferation and sustained induction of growth arrest and apoptosis. Bpv(pic) significantly decreases cell migration/invasion and anchorage-independent growth in soft agarose. Within cells, Bpv(pic) functions by attenuating CDC25A activity, and by decreasing the expression of multiple protein tyrosine phosphatases, DNA repair genes, microtubule-associated genes, such as PLK1, AURKA and HDAC6, and conversely augmenting the expression of proapoptotic mediators such as BAK, AIFM and CTSL1. CONCLUSION Collectively, our data strongly suggest novel evidence of Bpv(pic) being a potent antineoplastic drug and a suitable alternative for the treatment of pediatric low-grade gliomas.
Collapse
Affiliation(s)
- Norbert Fonya Ajeawung
- Department of Pediatrics, Laval University, 2705 Boulevard Laurier, Local RC9800, Québec, QC, G1V 4G2, Canada
| | | | | | | |
Collapse
|
27
|
Wang Y, Tian Y, Wan H, Li D, Wu W, Yin L, Jiang J, Wan W, Zhang L. Differences between brainstem gliomas in juvenile and adult rats. Oncol Lett 2013; 6:246-250. [PMID: 23946812 PMCID: PMC3742815 DOI: 10.3892/ol.2013.1319] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 04/16/2013] [Indexed: 11/25/2022] Open
Abstract
Clinical studies have shown that gliomas of the brainstem behave differently in children and adults. The aim of the present study was to compare and analyze the differences between these gliomas in juvenile and adult rats with regard to tumor growth, survival, pathology and magnetic resonance imaging (MRI). A total of 25 juvenile and 25 adult Wistar rats were divided into groups A (15 juvenile rats), B (10 juvenile rats), C (15 adult rats) and D (10 adult rats). The rats of groups A and C (experimental) were injected with glioma cells, while groups B and D (control) were injected with a physiological saline solution. Rat neurological signs, survival time, tumor size, hematoxylin and eosin (HE) staining and immunohistochemical staining for MMP-2, MMP-9 and β-catenin were compared. The survival time of group A was 19.47±2.232 days, whereas that of group C was 21.47±2.232 days (P<0.05). The tumor sizes were 4.55 and 4.62 mm (P>0.05) in groups A and C, respectively. HE and immunohistochemical staining revealed no differences between the groups. The results suggest that the growth patterns and invasiveness of brainstem gliomas may vary in children compared with adults due to the varied biological behaviors of the tumor cells.
Collapse
Affiliation(s)
- Yu Wang
- Department of Neurosurgery, Beijing Tian Tan Hospital, Capital Medical University, Beijing 100050, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mascelli S, Barla A, Raso A, Mosci S, Nozza P, Biassoni R, Morana G, Huber M, Mircean C, Fasulo D, Noy K, Wittemberg G, Pignatelli S, Piatelli G, Cama A, Garré ML, Capra V, Verri A. Molecular fingerprinting reflects different histotypes and brain region in low grade gliomas. BMC Cancer 2013; 13:387. [PMID: 23947815 PMCID: PMC3765921 DOI: 10.1186/1471-2407-13-387] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 08/06/2013] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND Paediatric low-grade gliomas (LGGs) encompass a heterogeneous set of tumours of different histologies, site of lesion, age and gender distribution, growth potential, morphological features, tendency to progression and clinical course. Among LGGs, Pilocytic astrocytomas (PAs) are the most common central nervous system (CNS) tumours in children. They are typically well-circumscribed, classified as grade I by the World Health Organization (WHO), but recurrence or progressive disease occurs in about 10-20% of cases. Despite radiological and neuropathological features deemed as classic are acknowledged, PA may present a bewildering variety of microscopic features. Indeed, tumours containing both neoplastic ganglion and astrocytic cells occur at a lower frequency. METHODS Gene expression profiling on 40 primary LGGs including PAs and mixed glial-neuronal tumours comprising gangliogliomas (GG) and desmoplastic infantile gangliogliomas (DIG) using Affymetrix array platform was performed. A biologically validated machine learning workflow for the identification of microarray-based gene signatures was devised. The method is based on a sparsity inducing regularization algorithm l₁l₂ that selects relevant variables and takes into account their correlation. The most significant genetic signatures emerging from gene-chip analysis were confirmed and validated by qPCR. RESULTS We identified an expression signature composed by a biologically validated list of 15 genes, able to distinguish infratentorial from supratentorial LGGs. In addition, a specific molecular fingerprinting distinguishes the supratentorial PAs from those originating in the posterior fossa. Lastly, within supratentorial tumours, we also identified a gene expression pattern composed by neurogenesis, cell motility and cell growth genes which dichotomize mixed glial-neuronal tumours versus PAs. Our results reinforce previous observations about aberrant activation of the mitogen-activated protein kinase (MAPK) pathway in LGGs, but still point to an active involvement of TGF-beta signaling pathway in the PA development and pick out some hitherto unreported genes worthy of further investigation for the mixed glial-neuronal tumours. CONCLUSIONS The identification of a brain region-specific gene signature suggests that LGGs, with similar pathological features but located at different sites, may be distinguishable on the basis of cancer genetics. Molecular fingerprinting seems to be able to better sub-classify such morphologically heterogeneous tumours and it is remarkable that mixed glial-neuronal tumours are strikingly separated from PAs.
Collapse
Affiliation(s)
- Samantha Mascelli
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Annalisa Barla
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| | - Alessandro Raso
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Sofia Mosci
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| | - Paolo Nozza
- Pathology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Roberto Biassoni
- Molecular Medicine Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Giovanni Morana
- Neuroradiology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Martin Huber
- Siemens AG, Corporate Technology, Freyeslebenstr. 1, 91058, Erlangen, Germany
| | - Cristian Mircean
- Siemens AG, Corporate Technology, Freyeslebenstr. 1, 91058, Erlangen, Germany
| | - Daniel Fasulo
- SCR - Siemens Corporate Research, Princeton, NJ, USA
| | - Karin Noy
- SCR - Siemens Corporate Research, Princeton, NJ, USA
| | | | - Sara Pignatelli
- Neuro-oncology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Gianluca Piatelli
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Armando Cama
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Maria Luisa Garré
- Neuro-oncology Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Valeria Capra
- Neurosurgery Unit, Istituto Giannina Gaslini, via G. Gaslini 5, 16147, Genoa, Italy
| | - Alessandro Verri
- DISI - Department of Computer Science, Università degli Studi di Genova, Via Dodecaneso, 35-16146, Genoa, Italy
| |
Collapse
|
29
|
Levesley J, Steele L, Taylor C, Sinha P, Lawler SE. ABT-263 enhances sensitivity to metformin and 2-deoxyglucose in pediatric glioma by promoting apoptotic cell death. PLoS One 2013; 8:e64051. [PMID: 23691145 PMCID: PMC3656874 DOI: 10.1371/journal.pone.0064051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/09/2013] [Indexed: 12/22/2022] Open
Abstract
Pediatric high grade glioma is refractory to conventional multimodal treatment, highlighting a need to develop novel efficacious therapies. We investigated tumor metabolism as a potential therapeutic target in a panel of diverse pediatric glioma cell lines (SF188, KNS42, UW479 and RES186) using metformin and 2-deoxyglucose. As a single agent, metformin had little effect on cell viability overall. SF188 cells were highly sensitive to 2-deoxyglucose however, combination of metformin with 2-deoxyglucose significantly reduced cell proliferation compared to either drug alone in all cell lines tested. In addition, the combination of the two agents was associated with a rapid decrease in cellular ATP and subsequent AMPK activation. However, increased cell death was only observed in select cell lines after prolonged exposure to the drug combination and was caspase independent. Anti-apoptotic BCL-2 family proteins have been indicated as mediators of resistance against metabolic stress. Therefore we sought to determine whether pharmacological inhibition of BCL-2/BCL-xL with ABT-263 could potentiate apoptosis in response to these agents. We found that ABT-263 increased sensitivity to 2-deoxyglucose and promoted rapid and extensive cell death in response to the combination of 2-deoxyglucose and metformin. Furthermore, cell death was inhibited by the pan-caspase inhibitor, z-VAD-FMK suggesting that ABT-263 potentiated caspase-dependent cell death in response to 2-deoxyglucose or its combination with metformin. Overall, these data provide support for the concept that targeting metabolic and anti-apoptotic pathways may be an effective therapeutic strategy in pediatric glioma.
Collapse
Affiliation(s)
- Jane Levesley
- Translational Neuro-Oncology Group, Leeds Institute of Molecular Medicine, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
- * E-mail: (JL); (SL)
| | - Lynette Steele
- Translational Neuro-Oncology Group, Leeds Institute of Molecular Medicine, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Claire Taylor
- Genomics Facility, Leeds Institute of Molecular Medicine, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Priyank Sinha
- Translational Neuro-Oncology Group, Leeds Institute of Molecular Medicine, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
| | - Sean E. Lawler
- Translational Neuro-Oncology Group, Leeds Institute of Molecular Medicine, University of Leeds, St James’s University Hospital, Leeds, United Kingdom
- * E-mail: (JL); (SL)
| |
Collapse
|
30
|
A phase II single-arm study of irinotecan in combination with temozolomide (TEMIRI) in children with newly diagnosed high grade glioma: a joint ITCC and SIOPE-brain tumour study. J Neurooncol 2013; 113:127-34. [DOI: 10.1007/s11060-013-1098-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 02/22/2013] [Indexed: 10/27/2022]
|
31
|
Abstract
World Health Organization (WHO) grade I astrocytomas include pilocytic astrocytoma (PA) and subependymal giant cell astrocytoma (SEGA). As technologies in pharmacologic neo-adjuvant therapy continue to progress and as molecular characteristics are progressively recognized as potential markers of both clinically significant tumor subtypes and response to therapy, interest in the biology of these tumors has surged. An updated review of the current knowledge of the molecular biology of these tumors is needed. We conducted a Medline search to identify published literature discussing the molecular biology of grade I astrocytomas. We then summarized this literature and discuss it in a logical framework through which the complex biology of these tumors can be clearly understood. A comprehensive review of the molecular biology of WHO grade I astrocytomas is presented. The past several years have seen rapid progress in the level of understanding of PA in particular, but the molecular literature regarding both PA and SEGA remains nebulous, ambiguous, and occasionally contradictory. In this review we provide a comprehensive discussion of the current understanding of the chromosomal, genomic, and epigenomic features of both PA and SEGA and provide a logical framework in which these data can be more readily understood.
Collapse
Affiliation(s)
- Nicholas F Marko
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
32
|
Gilheeney SW, Kieran MW. Differences in molecular genetics between pediatric and adult malignant astrocytomas: age matters. Future Oncol 2012; 8:549-58. [PMID: 22646770 DOI: 10.2217/fon.12.51] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The microscope - the classical tool for the investigation of cells and tissues - remains the basis for the classification of tumors throughout the body. Nowhere has this been more true than in the grading of astrocytomas. In spite of the fact that our parents warned us not to judge a book by its cover, we have continued to assume that adult and pediatric malignant gliomas that look the same, will have the same mutations, and thus respond to the same therapy. Rapid advances in molecular biology have permitted us the opportunity to go inside the cell and characterize the genetic events that underlie the true molecular heterogeneity of adult and pediatric brain tumors. In this paper, we will discuss some of the important clinical differences between pediatric and adult gliomas, with a focus on the molecular analysis of these different age groups.
Collapse
Affiliation(s)
- Stephen W Gilheeney
- Pediatric Neuro-Oncology, Dana-Farber Children's Hospital Cancer Center, Boston, MA, USA.
| | | |
Collapse
|
33
|
Gnekow AK, Falkenstein F, von Hornstein S, Zwiener I, Berkefeld S, Bison B, Warmuth-Metz M, Driever PH, Soerensen N, Kortmann RD, Pietsch T, Faldum A. Long-term follow-up of the multicenter, multidisciplinary treatment study HIT-LGG-1996 for low-grade glioma in children and adolescents of the German Speaking Society of Pediatric Oncology and Hematology. Neuro Oncol 2012; 14:1265-84. [PMID: 22942186 DOI: 10.1093/neuonc/nos202] [Citation(s) in RCA: 180] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Hirntumorstudien (HIT)-LGG-1996 protocol offered a comprehensive treatment strategy for pediatric patients with low-grade glioma (LGG), ie, observation, surgery, adjuvant radiotherapy, and chemotherapy to defer the start of irradiation in young children. In this current study, we sought to determine clinical factors for progression and survival. Between October 1, 1996 and March 31, 2004, 1031 patients were prospectively recruited into an observation arm (n = 668) and a nonsurgical arm stratifying 12 months of vincristine-carboplatin chemotherapy (n = 216) and conventional radiotherapy/brachytherapy (n = 147) in an age-dependent manner. Median patient age was 6.9 years; 28 patients had diencephalic syndrome, 44 had dissemination, and 108 had neurofibromatosis type 1(NF-1). Main tumor location was the supratentorial midline (40.4%), and the main histology was pilocytic astrocytoma (67.9%). Following a median observation of 9.3 years, 10-year overall survival (OS) was 0.94 and 10-year event-free survival (EFS) was 0.47. Ten-year progression-free survival was 0.62 following radiotherapy and 0.44 following chemotherapy. Sixty-one of 216 chemotherapy patients received radiotherapy 0.3-8.7 years after initial diagnosis. By multivariate analysis, diencephalic syndrome and incomplete resection were found to be unfavorable factors for OS and EFS, age ≥11 years for OS, and supratentorial midline location for EFS. Dissemination, age <1 year, and nonpilocytic histology were unfavorable factors for progression following radiotherapy (138 patients); and diencephalic syndrome, dissemination, and age ≥11 years were unfavorable factors following chemotherapy (210 patients). NF-1 patients and boys experienced prolonged tumor stabilization with chemotherapy. A nationwide multimodal treatment strategy is feasible for pediatric LGG. Extended follow-up yielded results comparable to single-institution series for the treatment groups. Three-quarters of surviving chemotherapy patients have not yet received radiation therapy. Infants with or without diencephalic syndrome and dissemination bear the highest risk for death and progression following diagnosis or treatment.
Collapse
Affiliation(s)
- Astrid K Gnekow
- Hospital for Children and Adolescents, Klinikum Augsburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
The term long-term epilepsy associated tumor (LEAT) encompasses lesions identified in patients investigated for long histories (often 2 years or more) of drug-resistant epilepsy. They are generally slowly growing, low grade, cortically based tumors, more often arising in younger age groups and in many cases exhibit neuronal in addition to glial differentiation. Gangliogliomas and dysembryoplastic neuroepithelial tumors predominate in this group. LEATs are further united by cyto-architectural changes that may be present in the adjacent cortex which have some similarities to developmental focal cortical dysplasias (FCD); these are now grouped as FCD type IIIb in the updated International League Against Epilepsy (ILAE) classification. In the majority of cases, surgical treatments are beneficial from both perspectives of managing the seizures and the tumor. However, in a minority, seizures may recur, tumors may show regrowth or recurrence, and rarely undergo anaplastic progression. Predicting and identifying tumors likely to behave less favorably are key objectives of the neuropathologist. With immunohistochemistry and modern molecular pathology, it is becoming increasingly possible to refine diagnostic groups. Despite this, some LEATs remain difficult to classify, particularly tumors with "non-specific" or diffuse growth patterns. Modification of LEAT classification is inevitable with the goal of unifying terminological criteria applied between centers for accurate clinico-pathological-molecular correlative data to emerge. Finally, establishing the epileptogenic components of LEAT, either within the lesion or perilesional cortex, will elucidate the cellular mechanisms of epileptogenesis, which in turn will guide optimal surgical management of these lesions.
Collapse
Affiliation(s)
- Maria Thom
- Department of Clinical and Experimental Epilepsy, UCL, Institute of Neurology, Queen Square, London, UK.
| | | | | |
Collapse
|
35
|
Kast RE, Lefranc F, Karpel-Massler G, Halatsch ME. Why dapsone stops seizures and may stop neutrophils' delivery of VEGF to glioblastoma. Br J Neurosurg 2012; 26:813-7. [PMID: 22551309 DOI: 10.3109/02688697.2012.674577] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Lopez-Gomez et al. recently published remarkable but mechanistically unexplained empirical evidence that the old antibiotic dapsone has antiepileptic activity. We addressed the question "Why should a sulfone antibiotic reduce seizures?". We report here our conclusions based on data from past studies that seizures are associated with elevated interleukin-8 (IL-8) and that dapsone inhibits IL-8 release and function in several different clinical and experimental contexts. Diverse CNS insults cause an increase in CNS IL-8. Thus, the pro-inflammatory environment generated by increase IL-8 leads to a lower seizure threshold. Together this evidence indicates dapsone exerts anti-seizure activity by diminishing IL-8 signalling. Since IL-8 is clearly upregulated in glioblastoma and contributes to the florid angiogenesis of that disease, and since interference with IL-8 function has been shown to inhibit glioblastoma invasion and growth in several experimental models, and dapsone has been repeatedly been shown to clinically inhibit IL-8 function when used to treat human neutrophilic dermatoses, we believe that dapsone thereby reduces seizures by countering IL-8 function and may similarly retard glioblastoma growth by such anti-IL-8 function.
Collapse
Affiliation(s)
- R E Kast
- Department of Psychiatry, University of Vermont, Burlington, VT 05401, USA.
| | | | | | | |
Collapse
|
36
|
Kast RE, Halatsch ME. Matrix Metalloproteinase-2 and -9 in Glioblastoma: A Trio of Old Drugs—Captopril, Disulfiram and Nelfinavir—Are Inhibitors with Potential as Adjunctive Treatments in Glioblastoma. Arch Med Res 2012; 43:243-7. [DOI: 10.1016/j.arcmed.2012.04.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/20/2012] [Indexed: 02/08/2023]
|
37
|
Ruge MI, Simon T, Suchorska B, Lehrke R, Hamisch C, Koerber F, Maarouf M, Treuer H, Berthold F, Sturm V, Voges J. Stereotactic brachytherapy with iodine-125 seeds for the treatment of inoperable low-grade gliomas in children: long-term outcome. J Clin Oncol 2011; 29:4151-9. [PMID: 21969508 DOI: 10.1200/jco.2011.37.3381] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Resection is generally considered the gold standard for treatment of low-grade (WHO grades I and II) gliomas (LGGs) in childhood. However, approximately 30% to 50% of these tumors are inoperable because of their localization in highly eloquent brain areas. A few reports have suggested stereotactic brachytherapy (SBT) with implantation of iodine-125 ((125)I) seeds as a safe and effective local treatment alternative. This single-center study provides a summary of the long-term outcome after SBT in one of the largest reported patient series. PATIENTS AND METHODS All pediatric patients treated with SBT ((125)I seeds; cumulative therapeutic dose 50-65 Gy within 9 months) by our group for LGG with follow-up of more than 6 months were included. Clinical and radiologic outcome, time to progression, and overall survival were evaluated. Prognostic factors (age, sex, Karnofsky performance score, tumor volume, and histology) for survival and disease progression were investigated. RESULTS In all, 147 of 160 pediatric patients treated with SBT (from 1982 through 2009) were analyzed in detail. Procedure-related mortality was zero, and the 30-day morbidity was transient and low (5.4%). Survival rates at 5 and 10 years were 93%, and 82%, respectively, with no significant difference between WHO grades I and II tumors (median follow-up, 67.1 ± 57.7 months). Twenty-one (14.8%) of 147 patients presented with tumor relapse. The remaining 126 patients revealed complete response in 24.6%, partial response in 31.0%, and stable disease in 29.6%. Neurologic status improved (57.8%) or remained stable (23.0%). None of the evaluated factors had significant impact on the study's end points except tumor volume more than 15 mL, which caused significantly higher rates of tumor recurrence (P < .05). CONCLUSION We demonstrate that SBT represents a safe, minimally invasive, and highly effective local treatment option for pediatric patients with inoperable LGG WHO grades I and II.
Collapse
|
38
|
Three paths to better tyrosine kinase inhibition behind the blood-brain barrier in treating chronic myelogenous leukemia and glioblastoma with imatinib. Transl Oncol 2011; 3:13-5. [PMID: 20165690 DOI: 10.1593/tlo.09280] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Revised: 09/28/2009] [Accepted: 10/02/2009] [Indexed: 12/24/2022] Open
Abstract
Chronic myelogenous leukemia (CML) can be controlled for years with the tyrosine kinase inhibitor imatinib but because imatinib poorly penetrates the blood-brain barrier (BBB), on occasion, the CML clone will thrive and evolve to an accelerated phase in the resulting imatinib sanctuary within the central nervous system. In this, CML resembles glioblastoma in that imatinib, which otherwise may be effective, cannot get to the tumor. Although a common street drug of abuse, methamphetamine is Food and Drug Administration-approved and marketed as a pharmaceutical drug to treat attention-deficit disorders. It has shown the ability to open the BBB in rodents. We have some clinical hints that it may do so in humans as well. This short note presents three new points potentially leading to better tyrosine kinase inhibition behind the BBB: 1) Pharmaceutical methamphetamine may have a useful role in treating both CML and glioblastoma by allowing higher imatinib concentrations behind the BBB. 2) The old antidepressant and monoamine oxidase inhibitor selegiline, used to treat Parkinson disease, is catabolized to methamphetamine. Selegiline, as a nonscheduled drug,may therefore be an easier way to open the BBB, allowing more effective chemotherapy with tyrosine kinases. 3) Dasatinib is a tyrosine kinase inhibitor with a spectrum of inhibition only partially overlapping that of imatinib and a mechanism of tyrosine kinase inhibition that is different from that of imatinib. The two should be additive. In addition, dasatinib crosses the BBB poorly, and it can therefore be expected to benefit from methamphetamine-assisted entry.
Collapse
|
39
|
Lo HW. Targeting Ras-RAF-ERK and its interactive pathways as a novel therapy for malignant gliomas. Curr Cancer Drug Targets 2011; 10:840-8. [PMID: 20718706 DOI: 10.2174/156800910793357970] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 08/17/2010] [Indexed: 11/22/2022]
Abstract
Malignant gliomas are the most common and the deadliest brain malignancies in adults. Despite the lack of a complete understanding of the biology of these tumors, significant advances have been made in the past decades. One of the key discoveries made in the area of malignant gliomas is that these tumors can be induced and maintained by aberrant signaling networks. In this context, the Ras pathway has been extensively exploited, from both basic and translational perspectives. Although somatic oncogenic mutations of Ras genes are frequent in several cancer types, early investigations on gliomas revealed disappointing facts that the Ras mutations are nearly absent in malignant gliomas and that the BRAF mutations are present in a very small percentage of gliomas. Therefore, the observed deregulation of the Ras-RAF-ERK signaling pathway in gliomas is attributed to its upstream positive regulators, including, EGFR and PDGFR known to be highly active in the majority of malignant gliomas. In contrast to the initial negative results on the somatic mutations of H-Ras, K-Ras and BRAF, recent breakthrough studies on pediatric low-grade astrocytomas uncovered genetic alterations of the BRAF gene involving copy number gains and rearrangements. The 7q34 rearrangements result in a novel in-frame KIAA1549:BRAF fusion gene that possesses constitutive BRAF kinase activity resembling oncogenic BRAF (V600E). In light of the earlier findings and recent breakthroughs, this review summarizes our current understanding of the Ras-RAF-ERK signaling pathway in gliomas and the outcome of preclinical and clinical studies that evaluated the efficacy of Ras-targeted therapy in malignant gliomas.
Collapse
Affiliation(s)
- Hui-Wen Lo
- Division of Surgical Sciences, Department of Surgery, Duke University School of Medicine, Duke Comprehensive Cancer Center and Duke Brain Tumor Center, 103 Research Drive, Durham, NC 27705, USA.
| |
Collapse
|