1
|
Ullah A, Ullah S, Waqas M, Khan M, Rehman NU, Khalid A, Jan A, Aziz S, Naeem M, Halim SA, Khan A, Al-Harrasi A. Novel Natural Inhibitors for Glioblastoma by Targeting Epidermal Growth Factor Receptor and Phosphoinositide 3-kinase. Curr Med Chem 2024; 31:6596-6613. [PMID: 38616761 DOI: 10.2174/0109298673293279240404080046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/12/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND/AIM Glioblastoma is an extensively malignant neoplasm of the brain that predominantly impacts the human population. To address the challenge of glioblastoma, herein, we have searched for new drug-like candidates by extensive computational and biochemical investigations. METHODS Approximately 950 compounds were virtually screened against the two most promising targets of glioblastoma, i.e., epidermal growth factor receptor (EGFR) and phosphoinositide 3-kinase (PI3K). Based on highly negative docking scores, excellent binding capabilities and good pharmacokinetic properties, eight and seven compounds were selected for EGFR and PI3K, respectively. RESULTS Among those hits, four natural products (SBEH-40, QUER, QTME-12, and HCFR) exerted dual inhibitory effects on EGFR and PI3K in our in-silico analysis; therefore, their capacity to suppress the cell proliferation was assessed in U87 cell line (type of glioma cell line). The compounds SBEH-40, QUER, and QTME-12 exhibited significant anti-proliferative capability with IC50 values of 11.97 ± 0.73 μM, 28.27 ± 1.52 μM, and 22.93 ± 1.63 μM respectively, while HCFR displayed weak inhibitory potency (IC50 = 74.97 ± 2.30 μM). CONCLUSION This study has identified novel natural products that inhibit the progression of glioblastoma; however, further examinations of these molecules are required in animal and tissue models to better understand their downstream targeting mechanisms.
Collapse
Affiliation(s)
- Atta Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Saeed Ullah
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
- Department of Biotechnology and Genetic Engineering, Hazara University, Mansehra, Pakistan
| | - Majid Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Najeeb Ur Rehman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia
| | - Afnan Jan
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Shahkaar Aziz
- Institute of Biotechnology and Genetic Engineering, The University of Agriculture, Peshawar 25130, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Sobia Ahsan Halim
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ajmal Khan
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat-ul-Mouz 616, Nizwa, Sultanate of Oman
| |
Collapse
|
2
|
Ahamed A, Hasan M, Samanta A, Alam SSM, Jamil Z, Ali S, Hoque M. Prospective pharmacological potential of cryptotanshinone in cancer therapy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 9:100308. [DOI: 10.1016/j.prmcm.2023.100308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
3
|
Han J, Deng H, Xiong Y, Xia X, Bao C, Chen L, Zhao Q, Zhong S, Peng L, Zhong C. Reduced PTCH2 expression is associated with glioma development through its regulation of the PTEN/AKT signaling pathway. Biochem Biophys Res Commun 2022; 627:76-83. [PMID: 36027694 DOI: 10.1016/j.bbrc.2022.08.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/06/2022] [Accepted: 08/14/2022] [Indexed: 12/01/2022]
Abstract
Mutations in the human protein patched homolog (PTCH) gene have been demonstrated to be associated with cancer development in several types of malignancy. However, the underlying mechanism of PTCH-associated cancer development remains poorly understood, to the best of our knowledge. In the present study, the expression of PTCH2 in glioma tumor tissues from The Cancer Genome Atlas (TCGA) database and clinical patients with glioma were measured. Reduced expression levels of PTCH2 were observed in patients with glioma with poor prognose. In vitro, overexpression of PTCH2 significantly suppressed the proliferation and invasion of the glioma cell lines, LN229 and U87-MG. Mechanistically, PTCH2 upregulated the expression of tumor suppressor PTEN, thereby leading to the suppression of pro-survival AKT signals in glioma. Reduced expression of PTEN and enhanced expression of AKT promoted glioma development in vitro and in vivo. Blockade of PTCH2/AKT signals efficiently strengthened the anticancer effects of chemotherapy and prolonged the survival time in tumor-bearing mice, which provided a novel insight into potential treatment strategies for glioma in the clinic.
Collapse
Affiliation(s)
- Jizhong Han
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Huajiang Deng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Yu Xiong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Xiangguo Xia
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Changshun Bao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Qin Zhao
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Shunjie Zhong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China
| | - Chuanhong Zhong
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, China; Laboratory of Neurological Disease and Brain Function, Luzhou, 646000, China.
| |
Collapse
|
4
|
Wu T, Chen Y, Yang L, Wang X, Chen K, Xu D. Ribonuclease A Family Member 2 Promotes the Malignant Progression of Glioma Through the PI3K/Akt Signaling Pathway. Front Oncol 2022; 12:921083. [PMID: 35747836 PMCID: PMC9211777 DOI: 10.3389/fonc.2022.921083] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 05/04/2022] [Indexed: 11/13/2022] Open
Abstract
The treatment of patients with glioma still faces many difficulties. To further optimize treatment, it is necessary to identify more accurate markers as treatment targets and predict prognostic indicators. RNASE2 was identified as a differentially expressed gene (DEG) in glioma tissues using bioinformatics analysis. In glioma microarrays, 31.21% (54/173) and 68.79% (119/173) patients showed low and high RNASE2 protein expression levels, respectively. RNASE2 protein levels were considerably correlated with age, WHO grade, relapse, and death. Both mRNA and protein levels were associated with the overall survival of patients with glioma. To investigate the role of RNASE2, it was overexpressed or silenced in glioma cells. RNASE2 overexpression promoted cell proliferation, migration, and invasion. In addition, its overexpression promoted the growth of subcutaneous tumors and lung metastasis of glioma cells. Key protein levels in the PI3K/Akt signaling pathway were upregulated by RNASE2 overexpression. In contrast, RNASE2 knockdown had the opposite effects. Furthermore, LY294002 blocked the effects of RNASE2 on the cell function of glioma cells. In conclusion, RNASE2 is a novel marker associated with the diagnosis and prognosis of patients with glioma, and it promotes the malignant progression of gliomas through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Tingfeng Wu
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Tingfeng Wu,
| | - Yongxiu Chen
- Gynecology Department, Guangdong Women and Children Hospital, Guangzhou, China
| | - Liying Yang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangyu Wang
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ke'en Chen
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dianshuang Xu
- Department of Neurosurgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Agarwal S, Muniyandi P, Maekawa T, Kumar DS. Vesicular systems employing natural substances as promising drug candidates for MMP inhibition in glioblastoma: A nanotechnological approach. Int J Pharm 2018; 551:339-361. [PMID: 30236647 DOI: 10.1016/j.ijpharm.2018.09.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/16/2022]
Abstract
Glioblastoma multiforme (GBM), one of the most lethal Brain tumors, characterized by its high invasive nature and increased mortality rates forms a major bottleneck in transport of therapeutics across the Blood Brain Barrier (BBB). Matrix metalloproteinases (MMPs) are classified as enzymes, which are found to be up regulated in the Glioma tumor microenvironment and thus can be considered as a target for inhibition for curbing GBM. Many chemotherapeutics and techniques have been employed for inhibiting MMPs till now but all of them failed miserably and were withdrawn in clinical trials due to their inability in restricting the tumor growth or increasing the overall survival rates. Thus, the quest for finding the suitable MMP inhibitor is still on and there is a critical need for identification of novel compounds which can alter the BBB permeability, restrain tumor growth and prevent tumor recurrence. Currently, naturally derived substances are gaining widespread attention as tumor inhibitors and many studies have been reported by far highlighting their importance in restricting MMP expression thus serving as chemotherapeutics for cancer due to their minimal toxicity. These substances may serve as probable candidates for inhibiting MMP expression in GBM. However, targeting and delivering the inhibitor to its target site is an issue that needs to be overcome in order to attain maximum specificity and sustained release. The birth of nanotechnology served as a boon in delivering drugs to the most complicated areas thus paving way for Nano drug delivery. An efficient Nano carrier with ability to cross the BBB and competently kill the Glioma cells forms the prerequisite for GBM chemotherapy. Vesicular drug delivery systems are one such class of carriers, which have the capacity to release the drug at a predetermined rate at the target site thus minimizing any undesirable side effects. Exploiting vesicular systems as promising Nano drug carriers to formulate naturally derived substances, that can bypass the BBB and act as an inhibitor against MMPs in GBM is the main theme of this review.
Collapse
Affiliation(s)
- Srishti Agarwal
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Priyadharshni Muniyandi
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - Toru Maekawa
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan
| | - D Sakthi Kumar
- Bio Nano Electronics Research Center, Graduate School of Interdisciplinary New Science, Toyo University, Kawagoe, Saitama 350-8585, Japan.
| |
Collapse
|
6
|
Wang Y, Sun X, Ji K, Du L, Xu C, He N, Wang J, Liu Y, Liu Q. Sirt3-mediated mitochondrial fission regulates the colorectal cancer stress response by modulating the Akt/PTEN signalling pathway. Biomed Pharmacother 2018; 105:1172-1182. [PMID: 30021354 DOI: 10.1016/j.biopha.2018.06.071] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
Sirtuin-3 (Sirt3), a sub-family member of the nicotinamide adenine dinucleotide-dependent histone deacetylases, has been reported to be involved in mitochondrial oxidative stress regulation, mitochondrial calcium management, mitophagy activation, and mitochondrial energy metabolism. The aim of our study was to explore the functional role of Sirt3 in colorectal cancer stress, focusing particularly on its effects on mitochondrial fission. Our study demonstrated that Sirt3 was highly upregulated in colorectal cancer cells compared to normal rectal mucosa cells. However, the genetic ablation of Sirt3 reduced colorectal cancer cell viability, mobility and proliferation. At the molecular level, we found that Sirt3 knockdown suppressed the expression of adhesive factors and cyclins. Furthermore, Sirt3 deletion was also associated with mitochondrial membrane potential reduction, ROS overproduction, mPTP opening, mitochondrial pro-apoptotic upregulation, and caspase-9-related death programme activation. Furthermore, we determined that Sirt3 regulated the colorectal cancer stress response by modulating mitochondrial fission. The loss of Sirt3 triggered fatal mitochondrial fission by suppressing the Akt/PTEN pathway. Re-activation of the Akt/PTEN pathway combatted mitochondrial fission and promoted colorectal cancer mobility, survival, and growth. Altogether, these findings provide an additional rationale for the function of Sirt3 in supporting the growth and survival of colorectal cancer.
Collapse
Affiliation(s)
- Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Xiaohui Sun
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Liqing Du
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Chang Xu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Ningning He
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Jinhan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Yang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation and Molecular Nuclear Medicine, Tianjin 300192, China.
| |
Collapse
|
7
|
Ferrer VP, Moura Neto V, Mentlein R. Glioma infiltration and extracellular matrix: key players and modulators. Glia 2018; 66:1542-1565. [DOI: 10.1002/glia.23309] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 01/18/2018] [Accepted: 01/29/2018] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Rolf Mentlein
- Department of Anatomy; University of Kiel; Kiel Germany
| |
Collapse
|
8
|
Abstract
Background: The prognosis for most patients with primary brain tumors remains poor. Recent advances in molecular and cell biology have led to a greater understanding of molecular alterations in brain tumors. These advances are being translated into new therapies that will hopefully improve the prognosis for patients with brain tumors. Methods: We reviewed the literature on small molecule targeted agents and monoclonal antibodies used in brain tumor research and brain tumor clinical trials for the past 20 years. Results: Brain tumors commonly express molecular abnormalities. These alterations can lead to the activation of cell pathways involved in cell proliferation. This knowledge has led to interest in novel anti-brain-tumor therapies targeting key components of these pathways. Many drugs and monoclonal antibodies have been developed that modulate these pathways and are in various stages of testing. Conclusions: The use of targeted therapies against brain tumors promises to improve the prognosis for patients with brain tumors. However, as the molecular pathogenesis of brain tumors has not been linked to a single genetic defect or target, molecular agents may need to be used in combinations or in tandem with cytotoxic agents. Further study of these agents in well-designed cooperative clinical trials is needed.
Collapse
Affiliation(s)
- Nicholas Butowski
- Department of Neurological Surgery, University of California, San Francisco, CA 94143-0350, USA
| | | |
Collapse
|
9
|
Nochioka K, Okuda H, Tatsumi K, Morita S, Ogata N, Wanaka A. Hedgehog Signaling Components Are Expressed in Choroidal Neovascularization in Laser-induced Retinal Lesion. Acta Histochem Cytochem 2016; 49:67-74. [PMID: 27239075 PMCID: PMC4858541 DOI: 10.1267/ahc.15036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 02/22/2016] [Indexed: 12/21/2022] Open
Abstract
Choroidal neovascularization is one of the major pathological changes in age-related macular degeneration, which causes devastating blindness in the elderly population. The molecular mechanism of choroidal neovascularization has been under extensive investigation, but is still an open question. We focused on sonic hedgehog signaling, which is implicated in angiogenesis in various organs. Laser-induced injuries to the mouse retina were made to cause choroidal neovascularization. We examined gene expression of sonic hedgehog, its receptors (patched1, smoothened, cell adhesion molecule down-regulated by oncogenes (Cdon) and biregional Cdon-binding protein (Boc)) and downstream transcription factors (Gli1-3) using real-time RT-PCR. At seven days after injury, mRNAs for Patched1 and Gli1 were upregulated in response to injury, but displayed no upregulation in control retinas. Immunohistochemistry revealed that Patched1 and Gli1 proteins were localized to CD31-positive endothelial cells that cluster between the wounded retina and the pigment epithelium layer. Treatment with the hedgehog signaling inhibitor cyclopamine did not significantly decrease the size of the neovascularization areas, but the hedgehog agonist purmorphamine made the areas significantly larger than those in untreated retina. These results suggest that the hedgehog-signaling cascade may be a therapeutic target for age-related macular degeneration.
Collapse
Affiliation(s)
- Katsunori Nochioka
- Department of Ophthalmology, Nara Medical University Faculty of Medicine
| | - Hiroaki Okuda
- Department of Anatomy and Neuroscience, Nara Medical University Faculty of Medicine
| | - Kouko Tatsumi
- Department of Anatomy and Neuroscience, Nara Medical University Faculty of Medicine
| | - Shoko Morita
- Department of Anatomy and Neuroscience, Nara Medical University Faculty of Medicine
| | - Nahoko Ogata
- Department of Ophthalmology, Nara Medical University Faculty of Medicine
| | - Akio Wanaka
- Department of Anatomy and Neuroscience, Nara Medical University Faculty of Medicine
| |
Collapse
|
10
|
Kang JH, Adamson C. Novel chemotherapeutics and other therapies for treating high-grade glioma. Expert Opin Investig Drugs 2015; 24:1361-79. [PMID: 26289791 DOI: 10.1517/13543784.2015.1048332] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Despite extensive research, high-grade glioma (HGG) remains a dire diagnosis with no change in the standard of care in almost a decade. However, recent advancements uncovering molecular biomarkers of brain tumors and tumor-specific antigens targeted by immunotherapies provide opportunities for novel personalized treatment regimens to improve survival. AREAS COVERED In this review, the authors provide a comprehensive overview of recent therapeutic advancements in HGG. Furthermore, they describe new molecular biomarkers and molecular classifications, in addition to updated research on bevacizumab, targeted molecular therapies, immunotherapy and alternative delivery methods that overcome the blood-brain barrier to reach the target tumor tissue. Challenges regarding each therapy are also outlined. The authors also provide some insight into a novel non-chemotherapeutic treatment for malignant glioma, NovoTTFA, as well as a summary of current treatment options for recurrence. EXPERT OPINION Current research for treating malignant gliomas are paving the path to personalized therapy, including immunotherapy, that involve integrated genomic and histolopathologic data, as well as a multi-modal treatment regimen. Immunotherapy will potentially be the next addition to the current standard of care, specialized to the antigens presented on the tumors. The results of the current trials of multi-antigen vaccines are eagerly anticipated.
Collapse
Affiliation(s)
- Jennifer H Kang
- a 1 Duke University School of Medicine , Box 3807, Durham, NC, USA
| | - Cory Adamson
- b 2 Director, Molecular Neuro-oncology Lab, Duke Medical Center , DUMC Box 3807, Durham, NC, USA.,c 3 Chief of Neurosurgery, Durham VA Medical Center , 508 Fulton Street, Durham, NC, USA +1 919 698 3152 ; .,d 4 Duke Medical Center , DUMC Box 3807, Durham, NC, USA
| |
Collapse
|
11
|
Phosphatase and Tensin Homologue: Novel Regulation by Developmental Signaling. JOURNAL OF SIGNAL TRANSDUCTION 2015; 2015:282567. [PMID: 26339505 PMCID: PMC4539077 DOI: 10.1155/2015/282567] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/06/2015] [Accepted: 07/01/2015] [Indexed: 11/18/2022]
Abstract
Phosphatase and tensin homologue (PTEN) is a critical cell endogenous inhibitor of phosphoinositide signaling in mammalian cells. PTEN dephosphorylates phosphoinositide trisphosphate (PIP3), and by so doing PTEN has the function of negative regulation of Akt, thereby inhibiting this key intracellular signal transduction pathway. In numerous cell types, PTEN loss-of-function mutations result in unopposed Akt signaling, producing numerous effects on cells. Numerous reports exist regarding mutations in PTEN leading to unregulated Akt and human disease, most notably cancer. However, less is commonly known about nonmutational regulation of PTEN. This review focuses on an emerging literature on the regulation of PTEN at the transcriptional, posttranscriptional, translational, and posttranslational levels. Specifically, a focus is placed on the role developmental signaling pathways play in PTEN regulation; this includes insulin-like growth factor, NOTCH, transforming growth factor, bone morphogenetic protein, wnt, and hedgehog signaling. The regulation of PTEN by developmental mediators affects critical biological processes including neuronal and organ development, stem cell maintenance, cell cycle regulation, inflammation, response to hypoxia, repair and recovery, and cell death and survival. Perturbations of PTEN regulation consequently lead to human diseases such as cancer, chronic inflammatory syndromes, developmental abnormalities, diabetes, and neurodegeneration.
Collapse
|
12
|
Garg T, Bhandari S, Rath G, Goyal AK. Current strategies for targeted delivery of bio-active drug molecules in the treatment of brain tumor. J Drug Target 2015; 23:865-87. [PMID: 25835469 DOI: 10.3109/1061186x.2015.1029930] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Brain tumor is one of the most challenging diseases to treat. The major obstacle in the specific drug delivery to brain is blood-brain barrier (BBB). Mostly available anti-cancer drugs are large hydrophobic molecules which have limited permeability via BBB. Therefore, it is clear that the protective barriers confining the passage of the foreign particles into the brain are the main impediment for the brain drug delivery. Hence, the major challenge in drug development and delivery for the neurological diseases is to design non-invasive nanocarrier systems that can assist controlled and targeted drug delivery to the specific regions of the brain. In this review article, our major focus to treat brain tumor by study numerous strategies includes intracerebral implants, BBB disruption, intraventricular infusion, convection-enhanced delivery, intra-arterial drug delivery, intrathecal drug delivery, injection, catheters, pumps, microdialysis, RNA interference, antisense therapy, gene therapy, monoclonal/cationic antibodies conjugate, endogenous transporters, lipophilic analogues, prodrugs, efflux transporters, direct conjugation of antitumor drugs, direct targeting of liposomes, nanoparticles, solid-lipid nanoparticles, polymeric micelles, dendrimers and albumin-based drug carriers.
Collapse
Affiliation(s)
| | - Saurav Bhandari
- b Department of Quality Assurance , ISF College of Pharmacy , Moga , Punjab , India
| | | | | |
Collapse
|
13
|
Singh AR, Joshi S, George E, Durden DL. Anti-tumor effect of a novel PI3-kinase inhibitor, SF1126, in (12) V-Ha-Ras transgenic mouse glioma model. Cancer Cell Int 2014; 14:105. [PMID: 25425962 PMCID: PMC4243316 DOI: 10.1186/s12935-014-0105-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 10/07/2014] [Indexed: 11/29/2022] Open
Abstract
Background Growth factor mediated activation of RAS-MAP-kinase and PI3-kinase-AKT pathways are critical for the pathogenesis of glioblastoma. The attenuation of PI3-kinase/AKT signaling will be effective in regulating the tumorigenic phenotypes of the glioma cells. Methods Glioma cells derived from the brain of the 12 V-Ha-Ras transgenic mice were used to study the effect of PI-3 kinase inhibitor SF1126 on activation of AKT and ERK signaling, proliferation, vitronectin mediated migration and changes in the distribution of cortical actin on vitronectin in the glioma cells in vitro. The anti-tumor effects of SF1126 were also tested in vivo using pre-established tumors (subcutaneous injection of the glioma cells from 12 V-Ha-Ras transgenic mice) in a mouse xenograft model. Results Our results demonstrate that treatment of LacZ+, GFAP + and PCNA + 12 V-Ras Tg transformed astrocytes with SF1126 and LY294002 blocked the activation of AKT as well as EGF-induced phospho-ERK. Most notably, treatment of SF1126 blocked integrin-dependent migration in transwell and scratch assays and caused a significant change in the organization and distribution of cortical actin on vitronectin in the glioma cells. Moreover, SF1126 treatment inhibited in vitro proliferation of these cells and in vivo growth of pre-established subcutaneous tumors in a xenograft model. Conclusion The present study validate the potent anti-proliferative and anti-migratory activity of SF1126, in a V12 Ras oncogene driven glioma model and suggest that this effect is mediated potentially through a combined attenuation of PI3-kinase and MAP-kinase signaling pathways.
Collapse
Affiliation(s)
- Alok R Singh
- UCSD Department of Pediatrics, Moores UCSD Cancer Center, University of California School of Medicine, San Diego, CA 92093 USA
| | - Shweta Joshi
- UCSD Department of Pediatrics, Moores UCSD Cancer Center, University of California School of Medicine, San Diego, CA 92093 USA
| | | | - Donald L Durden
- UCSD Department of Pediatrics, Moores UCSD Cancer Center, University of California School of Medicine, San Diego, CA 92093 USA ; Division of Pediatric Hematology-Oncology, UCSD Rady Children's Hospital, La Jolla, CA USA
| |
Collapse
|
14
|
Herzog S, Fink MA, Weitmann K, Friedel C, Hadlich S, Langner S, Kindermann K, Holm T, Böhm A, Eskilsson E, Miletic H, Hildner M, Fritsch M, Vogelgesang S, Havemann C, Ritter CA, Meyer zu Schwabedissen HE, Rauch B, Hoffmann W, Kroemer HK, Schroeder H, Bien-Möller S. Pim1 kinase is upregulated in glioblastoma multiforme and mediates tumor cell survival. Neuro Oncol 2014; 17:223-42. [PMID: 25155357 DOI: 10.1093/neuonc/nou216] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The current therapy for glioblastoma multiforme (GBM), the most aggressive and common primary brain tumor of adults, involves surgery and a combined radiochemotherapy that controls tumor progression only for a limited time window. Therefore, the identification of new molecular targets is highly necessary. Inhibition of kinases has become a standard of clinical oncology, and thus the oncogenic kinase Pim1 might represent a promising target for improvement of GBM therapy. METHODS Expression of Pim1 and associated signaling molecules was analyzed in human GBM samples, and the potential role of this kinase in patients' prognosis was evaluated. Furthermore, we analyzed the in vivo role of Pim1 in GBM cell growth in an orthotopic mouse model and examined the consequences of Pim1 inhibition in vitro to clarify underlying pathways. RESULTS In comparison with normal brain, a strong upregulation of Pim1 was demonstrated in human GBM samples. Notably, patients with short overall survival showed a significantly higher Pim1 expression compared with GBM patients who lived longer than the median. In vitro experiments with GBM cells and analysis of patients' GBM samples suggest that Pim1 regulation is dependent on epidermal growth factor receptor. Furthermore, inhibition of Pim1 resulted in reduced cell viability accompanied by decreased cell numbers and increased apoptotic cells, as seen by elevated subG1 cell contents and caspase-3 and -9 activation, as well as modulation of several cell cycle or apoptosis regulatory proteins. CONCLUSIONS Altogether, Pim1 could be a novel therapeutic target, which should be further analyzed to improve the outcome of patients with aggressive GBM.
Collapse
Affiliation(s)
- Susann Herzog
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Matthias Alexander Fink
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Kerstin Weitmann
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Claudius Friedel
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Stefan Hadlich
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Sönke Langner
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Katharina Kindermann
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Tobias Holm
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Andreas Böhm
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Eskil Eskilsson
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Hrvoje Miletic
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Markus Hildner
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Michael Fritsch
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Silke Vogelgesang
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Christoph Havemann
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Christoph Alexander Ritter
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Henriette Elisabeth Meyer zu Schwabedissen
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Bernhard Rauch
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Wolfgang Hoffmann
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Heyo Klaus Kroemer
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Henry Schroeder
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| | - Sandra Bien-Möller
- Department of Pharmacology/C_DAT (S.H., M.H., M.A.F., T.H., A.B., H.E.M.z.S., H.K.K., B.R., S.B-M.); Institute of Pathology (S.V.); Institute of Pharmacy (C.A.R.); Institute for Community Medicine (K.W., C.H., W.H.); Clinic of Neurosurgery (C.F., M.F., H.S.); Institute of Radiology and Neuroradiology, Universitätsmedizin Greifswald, Greifswald, Germany (S.H., K.K., S.L.); Department of Biomedicine, University of Bergen, Bergen, Norway (E.E., H.M.); Department of Pathology, Haukeland University Hospital, Bergen, Norway (E.E., H.M.)
| |
Collapse
|
15
|
Cytotoxic autophagy in cancer therapy. Int J Mol Sci 2014; 15:10034-51. [PMID: 24905404 PMCID: PMC4100138 DOI: 10.3390/ijms150610034] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 04/17/2014] [Accepted: 05/19/2014] [Indexed: 01/01/2023] Open
Abstract
Autophagy is a process of cellular self-digestion, whereby the cell degrades subcellular materials in order to generate energy and metabolic precursors in order to prolong survival, classically under conditions of nutrient deprivation. Autophagy can also involve the degradation of damaged or aged organelles, and misfolded or damaged proteins to eliminate these components that might otherwise be deleterious to cellular survival. Consequently, autophagy has generally been considered a prosurvival response. Many, if not most chemotherapeutic drugs and radiation also promote autophagy, which is generally considered a cytoprotective response, in that its inhibition frequently promotes apoptotic cells death. Furthermore, it has been shown that conventional chemotherapeutic drugs and radiation alone rarely induce a form of autophagy that leads to cell death. However, there are multiple examples in the literature where newer chemotherapeutic agents, drug combinations or drugs in combination with radiation promote autophagic cell death. This review will describe autophagic cell death induced in breast tumor cells, lung cancer cells as well as glioblastoma, demonstrating that it cannot be concluded that stress induced autophagy is, of necessity, cytoprotective in function.
Collapse
|
16
|
Abstract
Malignant gliomas comprise a small percentage of all cancers, but continue to cause disproportionate levels of morbidity and mortality. Despite decades of intensive effort from many disciplines--surgery, radiation oncology and medicine--they remain refractory to cure and, in most cases, even to prolonged treatment response. Comprehensive multidisciplinary treatment is well recognized as the optimal approach. While continued advances and refinement in both surgical and radiotherapy-based techniques are certain, medical therapies are expanding at a much more rapid rate. This is due, in large part, to an understanding of the molecular events that underlie cancer pathogenesis and improved laboratory techniques to manufacture and study molecules that influence this process. This review will focus on medical therapies in the treatment of malignant glioma, never losing sight of their place as one of several therapeutic modalities used to confront brain cancer.
Collapse
Affiliation(s)
- M Kelly Nicholas
- Department of Neurology, University of Chicago, 5801 South Ellis Ave., Chicago, IL 60637, USA.
| | | | | |
Collapse
|
17
|
Newton HB. Molecular neuro-oncology and the development of targeted therapeutic strategies for brain tumors Part 4: p53 signaling pathway. Expert Rev Anticancer Ther 2014; 5:177-91. [PMID: 15757449 DOI: 10.1586/14737140.5.1.177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Brain tumors are a diverse group of malignancies that remain refractory to conventional treatment approaches. Molecular neuro-oncology has now begun to clarify the transformed phenotype of brain tumors and identify oncogenic pathways that might be amenable to targeted therapy. Loss of the tumor suppressor gene p53 and its encoded protein are the most common genetic events in human cancer and are a frequent occurrence in brain tumors. p53 functions as a transcription factor and is responsible for the transactivation and repression of key genes involved in cell growth, apoptosis and the cell cycle. Mutation of the p53 gene or dysfunction of its signaling pathway are early events in the transformation process of astrocytic gliomas. The majority of mutations are missense and occur in the conserved regions of the gene, within exons 5 through 8. Molecular therapeutic strategies to normalize p53 signaling in cells with mutant p53 include pharmacologic rescue of mutant protein, gene therapy approaches, small-molecule agonists of downstream inhibitory genes, antisense approaches and oncolytic viruses. Other strategies include activation of normal p53 activity, inhibition of mdm2-mediated degradation of p53 and blockade of p53 nuclear export. Further development of targeted therapies designed to restore or enhance p53 function, and evaluation of these new agents in clinical trials, will be needed to improve survival and quality of life for patients with brain tumors.
Collapse
Affiliation(s)
- Herbert B Newton
- Dardinger Neuro-Oncology Center, Department of Neurology, Ohio State University Hospitals, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Abstract
Malignant gliomas are the most prevalent type of primary brain tumor in adults. Despite progress in brain tumor therapy, the prognosis of malignant glioma patients remains dismal. The median survival of patients with glioblastoma multiforme, the most common grade of malignant glioma, is 10-12 months. Conventional therapy of surgery, radiation and chemotherapy is largely palliative. Essentially, tumor recurrence is inevitable. Salvage treatments upon recurrence are palliative at best and rarely provide significant survival benefit. Therapies targeting the underlying molecular pathogenesis of brain tumors are urgently required. Common genetic abnormalities in malignant glioma specimens are associated with aberrant activation or suppression of cellular signal transduction pathways and resistance to radiation and chemotherapy. Several low molecular weight signal transduction inhibitors have been examined in preclinical and clinical malignant glioma trials. The efficacy of these agents as monotherapies has been modest, at best; however, small subsets of patients who harbor specific genetic changes in their tumors may display favorable clinical responses to defined small molecule inhibitors. Multitargeted kinase inhibitors or combinations of agents targeting different mitogenic pathways may overcome the resistance of tumors to single-agent targeted therapies. Well designed studies of small molecule kinase inhibitors will include assessment of safety, drug delivery, target inhibition and correlative biomarkers to define mechanisms of response or resistance to these agents. Predictive biomarkers will enrich for patients most likely to respond in future clinical trials. Additional clinical studies will combine novel targeted therapies with radiation, chemotherapies and immunotherapies.
Collapse
Affiliation(s)
- Sith Sathornsumetee
- The Preston Robert Tisch Brain Tumor Center Division of Neurosurgery/Neuro-Oncology, Duke University Medical Center, DUMC 3624, Durham, NC 27710, USA.
| | | |
Collapse
|
19
|
Abstract
Brain tumors remain a significant health problem. Advances in the biology of the blood-brain barrier are improving the ability of researchers to target therapeutic peptides, small molecules and other drugs to brain tumors. Simple methods to improve blood-brain barrier penetration include chemical modification, glycosylation and pegylation. Drug-delivery vehicles, such as nanoparticles and liposomes, are also under study. Targeting vectors include natural ligands (e.g., epidermal growth factor) or monoclonal antibodies to receptors (e.g., transferrin or insulin). Other vector-mediated delivery approaches involve the conjugation of a therapeutic peptide or protein with a targeting molecule that can induce transcytosis across blood-brain barrier endothelial cells. The most commonly used vectors are peptidomimetic antibodies to endothelial receptors, such as the transferrin and insulin receptors.
Collapse
Affiliation(s)
- Herbert B Newton
- Dardinger Neuro-oncology Center, Division of Neuro-oncology, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
20
|
Wen PY, Kesari S, Drappatz J. Malignant gliomas: strategies to increase the effectiveness of targeted molecular treatment. Expert Rev Anticancer Ther 2014; 6:733-54. [PMID: 16759164 DOI: 10.1586/14737140.6.5.733] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recently, there has been increasing interest in the use of targeted molecular agents for the treatment of malignant gliomas. These agents are generally well tolerated but have demonstrated only modest activity. In this article, the current status of targeted molecular agents for malignant gliomas will be reviewed and strategies to improve their effectiveness will be discussed.
Collapse
Affiliation(s)
- Patrick Y Wen
- Harvard Medical School, Dana-Farber/Brigham and Women's Cancer Center, SW430D, Boston, MA 02115, USA.
| | | | | |
Collapse
|
21
|
Penas-Prado M, Gilbert MR. Molecularly targeted therapies for malignant gliomas: advances and challenges. Expert Rev Anticancer Ther 2014; 7:641-61. [PMID: 17492929 DOI: 10.1586/14737140.7.5.641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The identification of molecular markers associated with tumor but not with normal tissue has allowed the development of highly specific, targeted therapies for the treatment of cancer. Over the last several years, tremendous advances in our understanding of the genetic and molecular changes involved in the progression of malignant gliomas have triggered a large effort in the development of targeted therapies to treat these tumors. However, to date only a modest clinical benefit, limited to subsets of patients, has been demonstrated. Furthermore, despite a high degree of target selectivity, the use of targeted therapies often has systemic toxicity. The reasons behind this limited clinical success are complex and include the intricacy of the signaling pathways in gliomas and the heterogeneity of the disease process, compounded by existing limitations in assessing the efficacy of these novel agents when conventional end points and clinical trial designs are utilized. However, despite these difficulties targeted therapies remain a very attractive avenue of treatment for malignant gliomas. Three basic approaches are needed to overcome the hurdles associated with targeted therapies: first, further development of genetic profiling techniques will help to better determine the genetic changes and molecular pathways involved in gliomas and will potentially allow the design of individualized therapies based on the genetic and molecular signature of each tumor. Second, there is a need for the development of better combination strategies (complementary targeted agents or targeted agents with chemotherapy drugs) directed towards disease heterogeneity. Third, we need to optimize the design of preclinical and clinical trials to obtain the maximum amount of information in the shortest period of time.
Collapse
Affiliation(s)
- Marta Penas-Prado
- The UT MD Anderson Cancer Center, Department of Neuro-Oncology, Houston, 77030 TX, USA.
| | | |
Collapse
|
22
|
Abstract
Glioblastoma multiforme represents the most common primary malignant tumor of the adult CNS. Unfortunately, the median survival after surgical intervention alone is less than 6 months and the addition of radiotherapy can extend this time to only 9 months. Consequently, efforts aimed at developing new therapies have focused on new treatment strategies that specifically target tumor cells and spare normal cells. One such modality, gene therapy, has shown promise in the spectrum of agents utilized against brain tumors. This review highlights the principles of gene therapy and discusses the results of recent clinical trials in which gene therapy has been employed against malignant brain tumors.
Collapse
Affiliation(s)
- Maciej S Lesniak
- Division of Neurological Surgery, The University of Chicago, Pritzker School of Medicine, 5841 S. Maryland Avenue, MC3026, Chicago, IL 60637, USA.
| |
Collapse
|
23
|
Cryptotanshinone inhibits human glioma cell proliferation by suppressing STAT3 signaling. Mol Cell Biochem 2013; 381:273-82. [PMID: 23740516 DOI: 10.1007/s11010-013-1711-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2013] [Accepted: 05/24/2013] [Indexed: 12/19/2022]
Abstract
Malignant gliomas (MGs) are among the most aggressive types of cancers in the human brain. Frequent tumor recurrence caused by a lack of effective therapeutic approaches results in a poor prognosis. Signal transducer and activator of transcription 3 (STAT3), an oncogenic protein, is constitutively activated in MGs and predicts a poor clinical outcome. STAT3 therefore is considered to be a promising target for the treatment of MGs. Cryptotanshinone (CTS), the main bioactive compound from the root of Salvia miltiorrhiza Bunge, has been reported to have various pharmacological effects. However, little is known about its function in MG cells. In this study, we evaluated the effect of CTS on the proliferation of human glioma cell lines (T98G and U87). Our results revealed that CTS significantly suppresses glioma cell proliferation. The phosphorylation of STAT3 Tyr705, but not Ser727, was inhibited by CTS, and STAT3 nuclear translocation was attenuated. Overexpression of constitutively active mutant STAT3C reversed the inhibitory effect of CTS, while knockdown STAT3 showed a similar inhibitory effect as CTS treatment. Following the downregulation of STAT3-regulated proteins cyclinD1 and survivin, cell cycle progression significantly arrested in G1/G0 phase. These results indicate that CTS may be a potential antiproliferation agent for the treatment of MGs and that its mechanism may be related to the inhibition of STAT3 signaling.
Collapse
|
24
|
Romano C, Schepis C. PTEN gene: a model for genetic diseases in dermatology. ScientificWorldJournal 2012; 2012:252457. [PMID: 22623890 PMCID: PMC3353286 DOI: 10.1100/2012/252457] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 01/04/2012] [Indexed: 01/04/2023] Open
Abstract
PTEN gene is considered one of the most mutated tumor suppressor genes in human cancer, and it's likely to become the first one in the near future. Since 1997, its involvement in tumor suppression has smoothly increased, up to the current importance. Germline mutations of PTEN cause the PTEN hamartoma tumor syndrome (PHTS), which include the past-called Cowden, Bannayan-Riley-Ruvalcaba, Proteus, Proteus-like, and Lhermitte-Duclos syndromes. Somatic mutations of PTEN have been observed in glioblastoma, prostate cancer, and brest cancer cell lines, quoting only the first tissues where the involvement has been proven. The negative regulation of cell interactions with the extracellular matrix could be the way PTEN phosphatase acts as a tumor suppressor. PTEN gene plays an essential role in human development. A recent model sees PTEN function as a stepwise gradation, which can be impaired not only by heterozygous mutations and homozygous losses, but also by other molecular mechanisms, such as transcriptional regression, epigenetic silencing, regulation by microRNAs, posttranslational modification, and aberrant localization. The involvement of PTEN function in melanoma and multistage skin carcinogenesis, with its implication in cancer treatment, and the role of front office in diagnosing PHTS are the main reasons why the dermatologist should know about PTEN.
Collapse
Affiliation(s)
- Corrado Romano
- Unit of Pediatrics and Medical Genetics, I.R.C.C.S. Associazione Oasi Maria Santissima, Troina, Italy
| | | |
Collapse
|
25
|
General and neurological complications of targeted therapy. HANDBOOK OF CLINICAL NEUROLOGY 2012; 105:937-45. [PMID: 22230543 DOI: 10.1016/b978-0-444-53502-3.00033-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
26
|
Nieto-Sampedro M, Valle-Argos B, Gómez-Nicola D, Fernández-Mayoralas A, Nieto-Díaz M. Inhibitors of Glioma Growth that Reveal the Tumour to the Immune System. Clin Med Insights Oncol 2011; 5:265-314. [PMID: 22084619 PMCID: PMC3201112 DOI: 10.4137/cmo.s7685] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Treated glioblastoma patients survive from 6 to 14 months. In the first part of this review, we describe glioma origins, cancer stem cells and the genomic alterations that generate dysregulated cell division, with enhanced proliferation and diverse response to radiation and chemotherapy. We review the pathways that mediate tumour cell proliferation, neo-angiogenesis, tumor cell invasion, as well as necrotic and apoptotic cell death. Then, we examine the ability of gliomas to evade and suppress the host immune system, exhibited at the levels of antigen recognition and immune activation, limiting the effective signaling between glioma and host immune cells.The second part of the review presents current therapies and their drawbacks. This is followed by a summary of the work of our laboratory during the past 20 years, on oligosaccharide and glycosphingolipid inhibitors of astroblast and astrocytoma division. Neurostatins, the O-acetylated forms of gangliosides GD1b and GT1b naturally present in mammalian brain, are cytostatic for normal astroblasts, but cytotoxic for rat C6 glioma cells and human astrocytoma grades III and IV, with ID50 values ranging from 200 to 450 nM. The inhibitors do not affect neurons or fibroblasts up to concentrations of 4 μM or higher.At least four different neurostatin-activated, cell-mediated antitumoral processes, lead to tumor destruction: (i) inhibition of tumor neovascularization; (ii) activation of microglia; (iii) activation of natural killer (NK) cells; (iv) activation of cytotoxic lymphocytes (CTL). The enhanced antigenicity of neurostatin-treated glioma cells, could be related to their increased expression of connexin 43. Because neurostatins and their analogues show specific activity and no toxicity for normal cells, a clinical trial would be the logical next step.
Collapse
Affiliation(s)
- Manuel Nieto-Sampedro
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Beatriz Valle-Argos
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Diego Gómez-Nicola
- Instituto Cajal de Neurobiología, CSIC, 28002 Madrid, Spain
- Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | | | | |
Collapse
|
27
|
Abstract
Patients with newly diagnosed glioblastoma multiforme should undergo a maximal tumor resection and then, whenever possible, should be entered into a clinical trial. The current standard of care consists of external beam irradiation, to a total of 60 Gy over 6 weeks, in combination with low-dose daily temozolomide, followed by at least six cycles of adjuvant temozolomide. If radiotherapy and a temozolomide-based adjuvant regimen fail, the most active treatment approach appears to be bevacizumab and irinotecan. Molecular therapy, with drugs targeting growth factor receptors and critical signal transduction pathway mediators, is also under active investigation.
Collapse
Affiliation(s)
- Herbert B Newton
- Herbert B. Newton, MD Dardinger Neuro-Oncology Center, Department of Neurology, Ohio State University Medical Center, 465 Means Hall, 1654 Upham Drive, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Uchida H, Arita K, Yunoue S, Yonezawa H, Shinsato Y, Kawano H, Hirano H, Hanaya R, Tokimura H. Role of sonic hedgehog signaling in migration of cell lines established from CD133-positive malignant glioma cells. J Neurooncol 2011; 104:697-704. [PMID: 21380601 DOI: 10.1007/s11060-011-0552-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Accepted: 02/18/2011] [Indexed: 01/11/2023]
Abstract
The sonic hedgehog (SHH) signaling pathway is essential for normal development and embryogenic morphogenesis. In malignant neoplasms its inappropriate activation correlates with tumorigenesis, proliferation, and migration. However, the role of SHH in infiltrative growth of glioblastoma remains to be elucidated. CD133 is a marker of tumor stem cells in glioblastoma, which are thought to play important roles in tumorigenesis, drug resistance, and tumor recurrence. We investigated the role of the SHH signaling pathway in migration of glioblastoma cell lines derived from CD133-positive cells. Two cell lines, GBM1 and GBM2, were established from CD133-positive cells sorted on an automagnetic cell separator from dispersed human glioblastoma cells. Both cell lines exhibited sphere-like growth in serum-free medium containing growth factor. Expression of patched (PTCH)-, a receptor of SHH, of smoothened (SMO)-, a 7 transmembrane receptor, and of GLI1- and GLI2, PTCH cascade signal proteins, was evaluated by reverse-transcription polymerase chain reaction (RT-PCR). The effects of recombinant SHH in the medium, and of knockdown of SMO-, GLI1- or GLI2 messenger RNA (mRNA) on the migratory ability of neoplastic cells were evaluated by scratch assays. RT-PCR revealed the presence of PTCH-, SMO-, GLI1-, and GLI2 mRNA in these cells. Their migratory ability was significantly enhanced (P < 0.05) by addition of recombinant SHH to the medium. Knockdown of SMO-, GLI1- or GLI2 mRNA resulted in significant decrease in the mobility of the neoplastic cells. Our study suggests that the SHH pathway plays an important role in the migratory ability of cells derived from CD133-positive human glioblastoma cells.
Collapse
Affiliation(s)
- Hiroyuki Uchida
- Department of Neurosurgery, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1, Sakuragaoka, Kagoshima, 890-8520, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Malignant gliomas are among the most devastating tumors for which conventional therapies have not significantly improved patient outcome. Despite advances in imaging, surgery, chemotherapy and radiotherapy, survival is still less than 2 years from diagnosis and more targeted therapies are urgently needed. Notch signaling is central to the normal and neoplastic development of the central nervous system, playing important roles in proliferation, differentiation, apoptosis and cancer stem cell regulation. Notch is also involved in the regulation response to hypoxia and angiogenesis, which are typical tumor and more specifically glioblastoma multiforme (GBM) features. Targeting Notch signaling is therefore a promising strategy for developing future therapies for the treatment of GBM. In this review we give an overview of the mechanisms of Notch signaling, its networking pathways in gliomas, and discuss its potential for designing novel therapeutic approaches.
Collapse
|
30
|
Abstract
Glioblastoma (GBM) is the most common primary tumor of the CNS in the adult. It is characterized by exponential growth and diffuse invasiveness. Among many different genetic alterations in GBM, e.g., mutations of PTEN, EGFR, p16/p19 and p53 and their impact on aberrant signaling have been thoroughly characterized. A major barrier to develop a common therapeutic strategy is founded on the fact that each tumor has its individual genetic fingerprint. Nonetheless, the PI3K pathway may represent a common therapeutic target to most GBM due to its central position in the signaling cascade affecting proliferation, apoptosis and migration. The read-out of blocking PI3K alone or in combination with other cancer pathways should mainly focus, besides the cytostatic effect, on cell death induction since sublethal damage may induce selection of more malignant clones. Targeting more than one pathway instead of a single agent approach may be more promising to kill GBM cells.
Collapse
|
31
|
Yang F, Brown C, Buettner R, Hedvat M, Starr R, Scuto A, Schroeder A, Jensen M, Jove R. Sorafenib induces growth arrest and apoptosis of human glioblastoma cells through the dephosphorylation of signal transducers and activators of transcription 3. Mol Cancer Ther 2010; 9:953-62. [PMID: 20371721 DOI: 10.1158/1535-7163.mct-09-0947] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Glioblastoma is the most common type of primary brain tumor and is rapidly progressive with few treatment options. Here, we report that sorafenib (< or =10 micromol/L) inhibited cell proliferation and induced apoptosis in two established cell lines (U87 and U251) and two primary cultures (PBT015 and PBT022) from human glioblastomas. The effects of sorafenib on these tumor cells were associated with inhibiting phosphorylated signal transducers and activators of transcription 3 (STAT3; Tyr705). Expression of a constitutively activated STAT3 mutant partially blocked the effects of sorafenib, consistent with a role for STAT3 inhibition in the response to sorafenib. Phosphorylated Janus-activated kinase (JAK)1 was inhibited in U87 and U251 cells, whereas phosphorylated JAK2 was inhibited in primary cultures. Sodium vanadate, a general inhibitor of protein tyrosine phosphatases, blocked the inhibition of phosphorylation of STAT3 (Tyr705) induced by sorafenib. These data indicate that the inhibition of STAT3 activity by sorafenib involves both the inhibition of upstream kinases (JAK1 and JAK2) of STAT3 and increased phosphatase activity. Phosphorylation of AKT was also reduced by sorafenib. In contrast, mitogen-activated protein kinases were not consistently inhibited by sorafenib in these cells. Two key cyclins (D and E) and the antiapoptotic protein Mcl-1 were downregulated by sorafenib in both cell lines and primary cultures. Our data suggest that inhibition of STAT3 signaling by sorafenib contributes to growth arrest and induction of apoptosis in glioblastoma cells. These findings provide a rationale for potential treatment of malignant gliomas with sorafenib. Mol Cancer Ther; 9(4); 953-62. (c)2010 AACR.
Collapse
Affiliation(s)
- Fan Yang
- Molecular Medicine, Beckman Research Institute, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
De Witt Hamer PC. Small molecule kinase inhibitors in glioblastoma: a systematic review of clinical studies. Neuro Oncol 2010; 12:304-16. [PMID: 20167819 PMCID: PMC2940593 DOI: 10.1093/neuonc/nop068] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 09/29/2009] [Indexed: 12/20/2022] Open
Abstract
The efficacy of small-molecule kinase inhibitors has recently changed standard clinical practice for several solid cancers. Glioblastoma is a solid cancer that universally recurs and unrelentingly results in death despite maximal surgery and radiotherapy with concomitant and adjuvant temozolomide. Several clinical studies using kinase inhibitors in glioblastoma have been reported. The present study systematically reviews the efficacy, toxicity, and tissue analysis of small-molecule kinase inhibitors in adult patients with glioblastoma as reported in published clinical studies and determines which kinases have been targeted by the inhibitors used in these studies. Publications were retrieved using a MEDLINE search and by screening meeting abstracts. A total of 60 studies qualified for inclusion, of which 25 were original reports. A total of 2385 glioblastoma patients receiving kinase inhibitors could be evaluated. The study designs included 2 phase III studies and 37 phase II studies. Extracted data included radiological response, progression-free survival, overall survival, toxicity, and biomarker analysis. The main findings were that (i) efficacy of small-molecule kinase inhibitors in clinical studies with glioblastoma patients does not yet warrant a change in standard clinical practice and (ii) 6 main kinase targets for inhibitors have been evaluated in these studies: EGFR, mTOR, KDR, FLT1, PKCbeta, and PDGFR.
Collapse
Affiliation(s)
- Philip C De Witt Hamer
- Neurosurgical Center Amsterdam, VU University Medical Center, De Boelelaan 1117, Amsterdam, The Netherlands.
| |
Collapse
|
33
|
Mure H, Matsuzaki K, Kitazato KT, Mizobuchi Y, Kuwayama K, Kageji T, Nagahiro S. Akt2 and Akt3 play a pivotal role in malignant gliomas. Neuro Oncol 2009; 12:221-32. [PMID: 20167810 DOI: 10.1093/neuonc/nop026] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Akt, one of the major downstream effectors of phosphatidylinositol 3-kinase, is hyper-expressed and activated in a variety of cancers including glioblastoma. However, the expression profiles of the Akt isoforms Akt1/PKBalpha, Akt2/PKBbeta, and Akt3/PKBgamma and their functional roles in malignant glioma are not well understood. Therefore, we examined the protein and mRNA expression patterns of Akt isoforms in tissues from human astrocytomas, glioblastomas, and non-neoplastic regions. We also explored the biological role of each Akt isoform in malignant glioma cells using RNA interference-mediated knock-down and the over-expression of plasmid DNA of each isoform. The expression of Akt1 protein and mRNA was similar in glioma and normal control tissues. Although the protein and mRNA level of Akt2 increased with the pathological grade of malignancy, the expression of Akt3 mRNA and protein decreased as the malignancy grade increased. In U87MG, T98G, and TGB cells, the down-regulation of Akt2 or Akt3 by RNA interference reduced the expression of the phosphorylated form of Bad, resulting in the induction of caspase-dependent apoptosis. Akt1 knock-down did not affect cell growth or survival. We first demonstrate that the over-expression of Akt2 or Akt3 down-regulated the expression of the other protein and that endogenous Akt3 protein showed high kinase activity in U87MG cells. Our data suggest that Akt2 and Akt3 play an important role in the viability of human malignant glioma cells. Targeting Akt2 and Akt3 may hold promise for the treatment of patients with gliomas.
Collapse
Affiliation(s)
- Hideo Mure
- Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, 3-18-15, Kuramoto-cho, Tokushima, Tokushima 770-8503, Japan.
| | | | | | | | | | | | | |
Collapse
|
34
|
Thaker NG, Pollack IF. Molecularly targeted therapies for malignant glioma: rationale for combinatorial strategies. Expert Rev Neurother 2009; 9:1815-36. [PMID: 19951140 PMCID: PMC2819818 DOI: 10.1586/ern.09.116] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Median survival of patients with malignant glioma (MG) from time of diagnosis is approximately 1 year, despite surgery, irradiation and conventional chemotherapy. Improving patient outcome relies on our ability to develop more effective therapies that are directed against the unique molecular aberrations within a patient's tumor. Such molecularly targeted therapies may provide novel treatments that are more effective than conventional chemotherapeutics. Recently developed therapeutic strategies have focused on targeting several core glioma signaling pathways, including pathways mediated by growth-factors, PI3K/Akt/PTEN/mTOR, Ras/Raf/MEK/MAPK and other vital pathways. However, given the molecular diversity, heterogeneity and diverging and converging signaling pathways associated with MG, it is unlikely that any single agent will have efficacy in more than a subset of tumors. Overcoming these therapeutic barriers will require multiple agents that can simultaneously inhibit these processes, providing a rationale for combination therapies. This review summarizes the currently implemented single-agent and combination molecularly targeted therapies for MG.
Collapse
Affiliation(s)
- Nikhil G Thaker
- Doris Duke Clinical Research Fellow, Departments of Neurosurgery, Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260 and 6 Oakwood Place, Voorhees, NJ 08043, USA Tel.: +1 856 392 4727 Fax: +1 412 692 5921
| | - Ian F Pollack
- Department of Neurosurgery, Children’s Hospital of Pittsburgh, University of Pittsburgh Brain Tumor Center, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, 3501 Fifth Avenue, University of Pittsburgh, Pittsburgh, PA 15213, USA Tel.: +1 412 692 5881 Fax: +1 412 692 5921
| |
Collapse
|
35
|
PKCα-MAPK/ERK-phospholipase A2 signaling is required for human melanoma-enhanced brain endothelial cell proliferation and motility. Microvasc Res 2009; 78:338-57. [DOI: 10.1016/j.mvr.2009.09.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2008] [Revised: 07/08/2009] [Accepted: 09/01/2009] [Indexed: 12/28/2022]
|
36
|
Laquintana V, Trapani A, Denora N, Wang F, Gallo JM, Trapani G. New strategies to deliver anticancer drugs to brain tumors. Expert Opin Drug Deliv 2009; 6:1017-32. [PMID: 19732031 DOI: 10.1517/17425240903167942] [Citation(s) in RCA: 137] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Malignant brain tumors are among the most challenging to treat and at present there are no uniformly successful treatment strategies. Standard treatment regimens consist of maximal surgical resection followed by radiotherapy and chemotherapy. The limited survival advantage attributed to chemotherapy is partially due to low CNS penetration of antineoplastic agents across the blood-brain barrier (BBB). OBJECTIVE The objective of this paper is to review recent approaches to delivering anticancer drugs into primary brain tumors. METHODS Both preclinical and clinical strategies to circumvent the BBB are considered that include chemical modification and colloidal carriers. CONCLUSION Analysis of the available data indicates that new approaches may be useful for CNS delivery, yet an appreciation of pharmacokinetic issues and improved knowledge of tumor biology will be needed to affect significantly drug delivery to the target site.
Collapse
|
37
|
Gonzalez J, de Groot J. Combination therapy for malignant glioma based on PTEN status. Expert Rev Anticancer Ther 2009; 8:1767-79. [PMID: 18983237 DOI: 10.1586/14737140.8.11.1767] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a central regulator of cell proliferation, migration, tumor growth, survival, angiogenesis and metabolism, contributing to the malignant phenotype of gliomas. Trials using targeted therapeutics against growth factor receptors and downstream signal mediators of the PI3K pathway have demonstrated only modest clinical benefit. Although recent clinical data suggests that malignant gliomas with PTEN are more likely to respond to EGF receptor inhibitors, gliomas have multiple concomitantly activated pathways, making them highly resistant to single-targeted therapy. This review discusses the importance of the PI3K pathway in glioma, the potential role of PTEN status in directing specific therapies, discusses clinical trial development of drug combinations to treat malignant gliomas and offers strategies for trial design that will be necessary to fully understand the successes and failures of current approaches to glioma therapy.
Collapse
Affiliation(s)
- Javier Gonzalez
- University of Texas Medical Branch, MD Anderson Cancer Center, Galveston, TX 77555-0144, USA.
| | | |
Collapse
|
38
|
Kim WY, Lee HY. Brain angiogenesis in developmental and pathological processes: mechanism and therapeutic intervention in brain tumors. FEBS J 2009; 276:4653-64. [PMID: 19664069 PMCID: PMC2847309 DOI: 10.1111/j.1742-4658.2009.07177.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Formation of new blood vessels is required for the growth and metastasis of all solid tumors. New blood vessels are established in tumors mainly through angiogenesis. Brain tumors in particular are highly angiogenic. Therefore, interventions designed to prevent angiogenesis may be effective at controlling brain tumors. Indeed, many recent findings from preclinical and clinical studies of antiangiogenic therapy for brain tumors have shown that it is a promising approach to managing this deadly disease, especially when combined with other cytotoxic treatments. In this minireview, we summarize the basic characteristics of brain tumor angiogenesis and the role of known angiogenic factors in regulating this angiogenesis, which may be targets of antiangiogenic therapy. We also discuss the current status of antiangiogenic therapy for brain tumors, the suggested mechanisms of this therapy and the limitations of this strategy.
Collapse
Affiliation(s)
- Woo-Young Kim
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|
39
|
Graf MR, Jia W, Johnson RS, Dent P, Mitchell C, Loria RM. Autophagy and the functional roles of Atg5 and beclin-1 in the anti-tumor effects of 3beta androstene 17alpha diol neuro-steroid on malignant glioma cells. J Steroid Biochem Mol Biol 2009; 115:137-45. [PMID: 19375507 DOI: 10.1016/j.jsbmb.2009.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 02/27/2009] [Accepted: 03/31/2009] [Indexed: 12/12/2022]
Abstract
In this study, we demonstrate that the anti-tumor activity of the neuro-steroid, 3beta androstene 17alpha diol (17alpha-AED) on malignant glioma cells is mediated by the induction of autophagy. 17alpha-AED can inhibit the proliferation an induce cell death of multiple, unrelated gliomas with an IC(50) between 8 and 25muM. 17alpha-AED treatment induced the formation of autophagosomes and acidic vesicular organelles in human malignant gliomas which was blocked by bafilomycin A1 or 3-methyladenine. Cleavage of microtubule-associated protein-light chain 3 (LC3), an essential step in autophagosome formation, was detected in human malignant glioma cells exposed to 17alpha-AED. In 17alpha-AED treated T98G glioma cells there was an increase in the autophagy related proteins Atg5 and beclin-1. Silencing of ATG5 or beclin-1 with small interfering RNA significantly reduced the incidence of autophagy in 17alpha-AED treated malignant gliomas and attenuated the cytotoxic effects of the neuro-steroid indicating that the induction of autophagy mediates the anti-glioma activity of 17alpha-AED rather than serving as a cyto-protective response. These results demonstrate that 17alpha-AED possesses significant anti-glioma activity when used at pharmacologically relevant concentrations in vitro and the cytotoxic effects are resultant from the induction of autophagy.
Collapse
Affiliation(s)
- Martin R Graf
- Department of Neurosurgery - Harold F. Young Neurosurgical Center and Massey Cancer Center, Virginia Commonwealth University Medical Center, PO Box 980631, Richmond, VA 23298-0631, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Djedid R, Kiss R, Lefranc F. Targeted therapy of glioblastomas: a 5-year view. ACTA ACUST UNITED AC 2009. [DOI: 10.2217/thy.09.12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
41
|
Lino M, Merlo A. Translating biology into clinic: the case of glioblastoma. Curr Opin Cell Biol 2009; 21:311-6. [PMID: 19217766 DOI: 10.1016/j.ceb.2008.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 12/29/2008] [Indexed: 12/31/2022]
Abstract
GBM, the most common and malignant primary tumor of the CNS, is characterized by exponential growth and diffuse invasiveness. Although the diverse causative genotypes that give rise to a inhomogeneous histological phenotype are well defined, effective therapy inducing tumor cell apoptosis has not been established so far. Following surgery, billions of invasive tumor cells remain to be targeted by systemic and local therapies. Targeting non-overlapping pathways, rather than a single agent approach, is more likely to be effective. The potential of local drug application has not been exploited yet. Systemically, novel drug combinations have to be developed that not only target key molecules at the signaling crossroads but also exploit energy demand and the epigenetic cancer program of GBM.
Collapse
Affiliation(s)
- Maddalena Lino
- Laboratory of Molecular Neuro-Oncology and Department of Neurosurgery, University Hospitals, Basel, Switzerland
| | | |
Collapse
|
42
|
Lefranc F, Rynkowski M, DeWitte O, Kiss R. Present and potential future adjuvant issues in high-grade astrocytic glioma treatment. Adv Tech Stand Neurosurg 2009; 34:3-35. [PMID: 19368079 DOI: 10.1007/978-3-211-78741-0_1] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Despite major advances in the management of malignant gliomas of which glioblastomas represent the ultimate grade of malignancy, they remain characterized by dismal prognoses. Glioblastoma patients have a median survival expectancy of only 14 months on the current standard treatment of surgical resection to the extent feasible, followed by adjuvant radiotherapy plus temozolomide, given concomitantly with and after radiotherapy. Malignant gliomas are associated with such dismal prognoses because glioma cells can actively migrate through the narrow extra-cellular spaces in the brain, often travelling relatively long distances, making them elusive targets for effective surgical management. Clinical and experimental data have demonstrated that invasive malignant glioma cells show a decrease in their proliferation rates and a relative resistance to apoptosis (type I programmed cell death) as compared to the highly cellular centre of the tumor, and this may contribute to their resistance to conventional pro-apoptotic chemotherapy and radiotherapy. Resistance to apoptosis results from changes at the genomic, transcriptional and post-transcriptional level of proteins, protein kinases and their transcriptional factor effectors. The PTEN/ PI3K/Akt/mTOR/NF-kappaB and the Ras/Raf/MEK/ERK signaling cascades play critical roles in the regulation of gene expression and prevention of apoptosis. Components of these pathways are mutated or aberrantly expressed in human cancer, notably glioblastomas. Monoclonal antibodies and low molecular-weight kinase inhibitors of these pathways are the most common classes of agents in targeted cancer treatment. However, most clinical trials of these agents as monotherapies have failed to demonstrate survival benefit. Despite resistance to apoptosis being closely linked to tumorigenesis, tumor cells can still be induced to die by non-apoptotic mechanisms such as necrosis, senescence, autophagy (type II programmed cell death) and mitotic catastrophe. Temozolomide brings significant therapeutic benefits in glioblastoma treatment. Part of temozolomide cytotoxic activity is exerted through pro-autophagic processes and also through the induction of late apoptosis. Autophagy, type II programmed cell death, represents an alternative mechanism to overcome, at least partly, the dramatic resistance of many cancers to pro-apoptotic-related therapies. Another way to potentially overcome apoptosis resistance is to decrease the migration of malignant glioma cells in the brain, which then should restore a level of sensitivity to pro-apoptotic drugs. Recent series of studies have supported the concept that malignant gliomas might be seen as an orchestration of cross-talks between cancer cells, microenvironment, vasculature and cancer stem cells. The present chapter focuses on (i) the major signaling pathways making glioblastomas resistant to apoptosis, (ii) the signaling pathways distinctly activated by pro-autophagic drugs as compared to pro-apoptotic ones, (iii) autophagic cell death as an alternative to combat malignant gliomas, (iv) the major scientific data already obtained by researchers to prove that temozolomide is actually a pro-autophagic and pro-apoptotic drug, (v) the molecular and cellular therapies and local drug delivery which could be used to complement conventional treatments, and a review of some of the currently ongoing clinical trials, (vi) the fact that reducing the levels of malignant glioma cell motility can restore pro-apoptotic drug sensitivity, (vii) the observation that inhibiting the sodium pump activity reduces both glioma cell proliferation and migration, (viii) the brain tumor stem cells as a target to complement conventional treatment.
Collapse
Affiliation(s)
- F Lefranc
- Department of Neurosurgery, Erasme University Hospital, Free University of Brussels, Brussels, Belgium
| | | | | | | |
Collapse
|
43
|
Marsh J, Mukherjee P, Seyfried TN. Akt-dependent proapoptotic effects of dietary restriction on late-stage management of a phosphatase and tensin homologue/tuberous sclerosis complex 2-deficient mouse astrocytoma. Clin Cancer Res 2008; 14:7751-62. [PMID: 19047102 DOI: 10.1158/1078-0432.ccr-08-0213] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Malignant astrocytomas exhibit constitutive Akt phosphorylation due to reduced phosphatase and tensin homologue (PTEN) tumor suppressor expression or to increased growth factor receptor tyrosine kinase activation. Many astrocytomas are also tuberous sclerosis complex 2 (TSC2) protein deficient and exhibit constitutive mammalian target of rapamycin (mTOR) activity. Astrocytomas harboring PTEN/Akt/TSC2 pathway mutations are dependent on glycolysis to satisfy their bioenergetic requirements. Therapies that disrupt energy homeostasis can potentially manage astrocytoma growth and progression. Although dietary restriction (DR) reduces glycolysis and manages early-stage astrocytoma growth, no prior studies have identified the mechanisms involved or determined if DR can also manage late-stage tumor growth. EXPERIMENTAL DESIGN The effects of a late-onset intermittent DR feeding paradigm were examined in adult C57BL/6J mice bearing the syngeneic CT-2A malignant astrocytoma grown orthotopically or subcutaneously. RESULTS In contrast to contralateral normal brain, CT-2A was PTEN/TSC2 protein deficient; exhibited constitutive Akt, mTOR, and BAD phosphorylation; and overexpressed insulin-like growth factor-I (IGF-I), IGF-I receptor, hypoxia-inducible transcription factor-1alpha (HIF-1alpha), type 1 glucose transporter protein (GLUT1), and pyruvate kinase. DR initiated 10 to 14 days after tumor implantation (late onset) reduced CT-2A growth, delayed malignant progression, and significantly extended survival. DR suppressed phosphorylation of Akt and BAD while reducing expression of IGF-I, HIF-1alpha, and GLUT1. DR also enhanced procaspase-9/procaspase-3 cleavage but had no effect mTOR phosphorylation. CONCLUSIONS Our findings indicate that IGF-I/Akt signaling is associated with the antiapoptotic and glycolytic phenotype of the CT-2A astrocytoma and that DR targets this pathway. Moreover, PTEN/TSC2 deficiency may impair adaptation to the DR-induced disruption of energy homeostasis, thus enhancing apoptosis. Our findings highlight the efficacy of late-onset DR in managing astrocytoma growth and suggest that DR may be an effective broad-spectrum inhibitor of Akt signaling in PTEN/TSC2-deficient astrocytomas.
Collapse
Affiliation(s)
- Jeremy Marsh
- Biology Department, Boston College, Chestnut Hill, Massachusetts 02467, USA
| | | | | |
Collapse
|
44
|
Fukai J, Yokote H, Yamanaka R, Arao T, Nishio K, Itakura T. EphA4 promotes cell proliferation and migration through a novel EphA4-FGFR1 signaling pathway in the human glioma U251 cell line. Mol Cancer Ther 2008; 7:2768-78. [PMID: 18790757 DOI: 10.1158/1535-7163.mct-07-2263] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The Eph receptor tyrosine kinases and their ephrin ligands form a unique cell-cell contact-mediated bidirectional signaling mechanism for regulating cell localization and organization. High expression of Eph receptors in a wide variety of human tumors indicates some roles in tumor progression, which makes these proteins potential targets for anticancer therapy. For this purpose, we did gene expression profiling for 47 surgical specimens of brain tumors including 32 high-grade glioma using a microarray technique. The analysis, focused on the receptor tyrosine kinases, showed that EphA4 mRNA in the tumors was 4-fold higher than in normal brain tissue. To investigate the biological significance of EphA4 overexpression in these tumors, we analyzed EphA4-induced phenotypic changes and the signaling mechanisms using human glioma U251 cells. EphA4 promoted fibroblast growth factor 2-mediated cell proliferation and migration accompanied with enhancement of fibroblast growth factor 2-triggered mitogen-activated protein kinase and Akt phosphorylation. In addition, active forms of Rac1 and Cdc42 increased in the EphA4-overexpressing cells. Furthermore, we found that EphA4 formed a heteroreceptor complex with fibroblast growth factor receptor 1 (FGFR1) in the cells and that the EphA4-FGFR1 complex potentiated FGFR-mediated downstream signaling. Thus, our results indicate that EphA4 plays an important role in malignant phenotypes of glioblastoma by enhancing cell proliferation and migration through accelerating a canonical FGFR signaling pathway.
Collapse
Affiliation(s)
- Junya Fukai
- Department of Neurological Surgery, Wakayama Medical University, Wakayama, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Expression of hypoxia inducible factor-1alpha in tumors of patients with glioblastoma multiforme and transitional meningioma. J Clin Neurosci 2008; 15:1036-42. [PMID: 18621534 DOI: 10.1016/j.jocn.2007.07.080] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2007] [Revised: 06/22/2007] [Accepted: 07/20/2007] [Indexed: 12/27/2022]
Abstract
Hypoxia-inducible factor-1 alpha (HIF-1alpha) is the major transcriptional factor involved in the adaptive response to hypoxia. The aim of this study was to assess HIF-1alpha in 22 patients with transitional meningioma (TM) and 26 patients with glioblastoma multiforme (GBM). HIF-1alpha was assessed using a commercially available enzyme-linked immunosorbent assay-based HIF-1 transcription factor assay. Levels of HIF-1alpha in TM and GBM were measured using optical density at 450nm, and median values were found to be 0.35 for TM and 0.37 OD for GBM, respectively. There was no statistically significant difference between the two types of tumor (p=0.264). These findings indicate that HIF-1alpha is elevated in both TM and GBM, suggesting that although hypoxia is one of the most important and powerful stimuli for HIF-1alpha elevation and consequently angiogenesis, other mechanisms may play roles in HIF-1alpha stimulation in benign brain tumors such as TM.
Collapse
|
46
|
Thorarinsdottir HK, Santi M, McCarter R, Rushing EJ, Cornelison R, Jales A, MacDonald TJ. Protein expression of platelet-derived growth factor receptor correlates with malignant histology and PTEN with survival in childhood gliomas. Clin Cancer Res 2008; 14:3386-94. [PMID: 18519768 PMCID: PMC2953416 DOI: 10.1158/1078-0432.ccr-07-1616] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We previously showed that overexpression of epidermal growth factor receptor (EGFR) is associated with malignant grade in childhood glioma. The objective of this study was to determine whether protein expression of EGFR or platelet-derived growth factor receptor (PDGFR) and their active signaling pathways are related to malignant histology, progression of disease, and worse survival. EXPERIMENTAL DESIGN Tissue microarrays were prepared from untreated tumors from 85 new glioma patients [22 high-grade gliomas (HGG) and 63 low-grade gliomas (LGG)] diagnosed at this institution from 1989 to 2004. Immunohistochemistry was used to assess total expression of EGFR, PDGFR beta, and PTEN and expression of phosphorylated EGFR, phosphorylated PDGFR alpha (p-PDGFR alpha), phosphorylated AKT, phosphorylated mitogen-activated protein kinase, and phosphorylated mammalian target of rapamycin. These results were correlated with clinicopathologic data, including extent of initial tumor resection, evidence of dissemination, tumor grade, proliferation index, and survival, as well as with Affymetrix gene expression profiles previously obtained from a subset of these tumors. RESULTS High expression of p-PDGFR alpha, EGFR, PDGFR beta, and phosphorylated EGFR was seen in 85.7%, 80.0%, 78.9%, and 47.4% of HGG and 40.0%, 87.1%, 41.7%, and 30.6% of LGG, respectively. However, high expression of p-PDGFR alpha and PDGFR beta was the only significant association with malignant histology (P = 0.031 and 0.005, respectively); only the loss of PTEN expression was associated with worse overall survival. None of these targets, either alone or in combination, was significantly associated with progression-free survival in either LGG or HGG. CONCLUSIONS High PDGFR protein expression is significantly associated with malignant histology in pediatric gliomas, but it does not represent an independent prognostic factor. Deficient PTEN expression is associated with worse overall survival in HGG.
Collapse
Affiliation(s)
| | | | - Robert McCarter
- Division of Biostatistics, Children’s National Medical Center
| | | | - Robert Cornelison
- Institute for Biomedical Sciences, George Washington University
- Cancer Genetics Branch, National Human Genome Research Institute, NIH, Bethesda, Maryland
| | | | - Tobey J. MacDonald
- Division of Hematology-Oncology, Children’s National Medical Center
- Institute for Biomedical Sciences, George Washington University
- Center for Cancer and Immunology Research, Children’s Research Institute, Washington, District of Columbia
| |
Collapse
|
47
|
Cdk5-mediated regulation of the PIKE-A-Akt pathway and glioblastoma cell invasion. Proc Natl Acad Sci U S A 2008; 105:7570-5. [PMID: 18487454 DOI: 10.1073/pnas.0712306105] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Isoform A of phosphatidylinositol 3-kinase enhancer (PIKE-A) is a newly identified prooncogenic factor that has been implicated in cancer cell growth. How PIKE-A activity is regulated in response to growth signal is poorly understood. Here, we demonstrate that cyclin dependent kinase 5 (Cdk5), a protein known to function mainly in postmitotic neurons, directly phosphorylates PIKE-A at Ser-279 in its GTPase domain in glioblastoma cells. This phosphorylation event stimulates PIKE-A GTPase activity and the activity of its downstream effector Akt. Growth signal activates Cdk5 and results in a Cdk5-dependent accumulation of phosphorylated PIKE-A and activation of Akt in the nucleus. Furthermore, PIKE-A phosphorylation and Cdk5 are increased in human glioblastoma specimens. Phosphorylation of PIKE-A by Cdk5 mediates growth factor-induced migration and invasion of human glioblastoma cells. Together, these findings identify PIKE as the first Cdk5 target in cancer cells, revealing a previously undescribed regulatory mechanism that mediates growth signal-induced activation of PIKE-A/Akt and tumor invasion.
Collapse
|
48
|
Abstract
BACKGROUND Malignant gliomas are amongst the most devastating and intractable of all cancers. The most common malignant glioma, glioblastoma multiforme (GBM), is associated with a median survival in the range of 12-15 months. Survival for patients with GBM has improved with the addition of temozolomide chemotherapy to post-operative radiotherapy. Further advances in the treatment of malignant glioma will hinge on the discovery of novel and likely targeted therapies with activity against these diseases. OBJECTIVE Review recent published experience using targeted therapeutics for malignant glioma. METHODS Key studies from a Medline review of targeted therapies for malignant glioma performed between 2000 and the present are summarised in this review. CONCLUSIONS Experience with targeted therapeutics for malignant glioma has been to date disappointing. These agents are generally well tolerated, but activity is limited. Novel therapeutics with activity against malignant gliomas must be identified to improve prognosis for patients with these diseases.
Collapse
Affiliation(s)
- Warren P Mason
- Princess Margaret Hospital, Department of Medicine, 610 University Avenue, Suite 18-717, Toronto, Ontario, M5G 2M9, Canada.
| |
Collapse
|
49
|
Nakano I, Masterman-Smith M, Saigusa K, Paucar AA, Horvath S, Shoemaker L, Watanabe M, Negro A, Bajpai R, Howes A, Lelievre V, Waschek JA, Lazareff JA, Freije WA, Liau LM, Gilbertson RJ, Cloughesy TF, Geschwind DH, Nelson SF, Mischel PS, Terskikh AV, Kornblum HI. Maternal embryonic leucine zipper kinase is a key regulator of the proliferation of malignant brain tumors, including brain tumor stem cells. J Neurosci Res 2008; 86:48-60. [PMID: 17722061 DOI: 10.1002/jnr.21471] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Emerging evidence suggests that neural stem cells and brain tumors regulate their proliferation via similar pathways. In a previous study, we demonstrated that maternal embryonic leucine zipper kinase (Melk) is highly expressed in murine neural stem cells and regulates their proliferation. Here we describe how MELK expression is correlated with pathologic grade of brain tumors, and its expression levels are significantly correlated with shorter survival, particularly in younger glioblastoma patients. In normal human astrocytes, MELK is only faintly expressed, and MELK knockdown does not significantly influence their growth, whereas Ras and Akt overexpressing astrocytes have up-regulated MELK expression, and the effect of MELK knockdown is more prominent in these transformed astrocytes. In primary cultures from human glioblastoma and medulloblastoma, MELK knockdown by siRNA results in inhibition of the proliferation and survival of these tumors. Furthermore, we show that MELK siRNA dramatically inhibits proliferation and, to some extent, survival of stem cells isolated from glioblastoma in vitro. These results demonstrate a critical role for MELK in the proliferation of brain tumors, including their stem cells, and suggest that MELK may be a compelling molecular target for treatment of high-grade brain tumors.
Collapse
Affiliation(s)
- Ichiro Nakano
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lefranc F, Kiss R. The sodium pump alpha1 subunit as a potential target to combat apoptosis-resistant glioblastomas. Neoplasia 2008; 10:198-206. [PMID: 18323016 PMCID: PMC2259449 DOI: 10.1593/neo.07928] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 12/17/2007] [Accepted: 12/18/2007] [Indexed: 12/21/2022]
Abstract
PURPOSE To review the involvement of the ion transporter Na+/K+-ATPase (NaK) in the migration and proliferation of glioma cells. Preliminary studies indicate that NaK alpha1 subunits seem to be upregulated in a proportion of glioblastomas but not in normal brain tissues. DESIGN The present review focuses on (1) the natural resistance of migrating malignant glioma cells to apoptosis, (2) autophagic cell death as an alternative to combat malignant gliomas, (3) the fact that reducing the levels of malignant glioma cell motility can restore proapoptotic drug sensitivity,and (4) on the observation that inhibiting the NaK activity reduces both glioma cell proliferation and migration. RESULTS The natural ligands of the NaK are the cardiotonic steroids. A hemisynthetic derivative of 2"-oxovoruscharin (UNBS1450), a novel cardenolide, displays unique structural features, making its binding affinity to NaK alpha subunits (including alpha1) 10 to 100 times higher than that of other cardenolides. UNBS1450 markedly decreases intracellular ATP concentration in glioma cells, disorganizes the actin cytoskeleton, and leads to autophagic cell death in NaK alpha1 over-expressing glioma cells. CONCLUSIONS Glioblastoma patients who do not respond to chemotherapy and whose tumors over-express NaK alpha1 subunits could benefit from a treatment using ligands with marked binding affinity for the NaK alpha1 subunit.
Collapse
Affiliation(s)
- Florence Lefranc
- Department of Neurosurgery, Erasme University Hospital,Université Libre de Bruxelles, Brussels, Belgium.
| | | |
Collapse
|