1
|
Singh S, Joshi V, Upadhyay A. Amyloids and brain cancer: molecular linkages and crossovers. Biosci Rep 2023; 43:BSR20230489. [PMID: 37335084 PMCID: PMC10548166 DOI: 10.1042/bsr20230489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/31/2023] [Accepted: 06/13/2023] [Indexed: 06/21/2023] Open
Abstract
Amyloids are high-order proteinaceous formations deposited in both intra- and extracellular spaces. These aggregates have tendencies to deregulate cellular physiology in multiple ways; for example, altered metabolism, mitochondrial dysfunctions, immune modulation, etc. When amyloids are formed in brain tissues, the endpoint often is death of neurons. However, interesting but least understood is a close connection of amyloids with another set of conditions in which brain cells proliferate at an extraordinary rate and form tumor inside brain. Glioblastoma is one such condition. Increasing number of evidence indicate a possible link between amyloid formation and depositions in brain tumors. Several proteins associated with cell cycle regulation and apoptotic pathways themselves have shown to possess high tendencies to form amyloids. Tumor suppressor protein p53 is one prominent example that mutate, oligomerize and form amyloids leading to loss- or gain-of-functions and cause increased cell proliferation and malignancies. In this review article, we present available examples, genetic links and common pathways that indicate that possibly the two distantly placed pathways: amyloid formation and developing cancers in the brain have similarities and are mechanistically intertwined together.
Collapse
Affiliation(s)
- Shalini Singh
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| | - Vibhuti Joshi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Biotechnology, School of Engineering and Applied Sciences, Bennett University, Greater Noida, Uttar Pradesh 201310, India
| | - Arun Upadhyay
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jheepasani, Jodhpur, Rajasthan 342001, India
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, U.S.A
| |
Collapse
|
2
|
El-Baba C, Ayache Z, Goli M, Hayar B, Kawtharani Z, Pisano C, Kobeissy F, Mechref Y, Darwiche N. The Antitumor Effect of the DNA Polymerase Alpha Inhibitor ST1926 in Glioblastoma: A Proteomics Approach. Int J Mol Sci 2023; 24:14069. [PMID: 37762371 PMCID: PMC10531065 DOI: 10.3390/ijms241814069] [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: 07/31/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Glioblastoma Multiforme (GBM) is the most aggressive form of malignant brain tumor. The median survival rate does not exceed two years, indicating an imminent need to develop novel therapies. The atypical adamantyl retinoid ST1926 induces apoptosis and growth inhibition in different cancer types. We have shown that ST1926 is an inhibitor of the catalytic subunit of DNA polymerase alpha (POLA1), which is involved in initiating DNA synthesis in eukaryotic cells. POLA1 levels are elevated in GBM versus normal brain tissues. Therefore, we studied the antitumor effects of ST1926 in several human GBM cell lines. We further explored the global protein expression profiles in GBM cell lines using liquid chromatography coupled with tandem mass spectrometry to identify new targets of ST1926. Low sub-micromolar concentrations of ST1926 potently decreased cell viability, induced cell damage and apoptosis, and reduced POLA1 protein levels in GBM cells. The proteomics profiles revealed 197 proteins significantly differentially altered upon ST1926 treatment of GBM cells involved in various cellular processes. We explored the differential gene and protein expression of significantly altered proteins in GBM compared to normal brain tissues.
Collapse
Affiliation(s)
- Chirine El-Baba
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
| | - Zeinab Ayache
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
| | - Mona Goli
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| | - Berthe Hayar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
| | - Zeinab Kawtharani
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
| | - Claudio Pisano
- Biogem, Institute of Molecular Biology and Genetics, 83031 Ariano Irpino, Italy;
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
- Department of Neurobiology, Center for Neurotrauma, Multiomics and Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX 79409, USA;
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut 1107 2020, Lebanon; (C.E.-B.); (Z.A.); (B.H.); (Z.K.); (F.K.)
| |
Collapse
|
3
|
Tluli O, Al-Maadhadi M, Al-Khulaifi AA, Akomolafe AF, Al-Kuwari SY, Al-Khayarin R, Maccalli C, Pedersen S. Exploring the Role of microRNAs in Glioma Progression, Prognosis, and Therapeutic Strategies. Cancers (Basel) 2023; 15:4213. [PMID: 37686489 PMCID: PMC10486509 DOI: 10.3390/cancers15174213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 09/10/2023] Open
Abstract
Gliomas, which arise from glial cells in the brain, remain a significant challenge due to their location and resistance to traditional treatments. Despite research efforts and advancements in healthcare, the incidence of gliomas has risen dramatically over the past two decades. The dysregulation of microRNAs (miRNAs) has prompted the creation of therapeutic agents that specially target them. However, it has been reported that they are involved in complex signaling pathways that contribute to the loss of expression of tumor suppressor genes and the upregulation of the expression of oncogenes. In addition, numerous miRNAs promote the development, progression, and recurrence of gliomas by targeting crucial proteins and enzymes involved in metabolic pathways such as glycolysis and oxidative phosphorylation. However, the complex interplay among these pathways along with other obstacles hinders the ability to apply miRNA targeting in clinical practice. This highlights the importance of identifying specific miRNAs to be targeted for therapy and having a complete understanding of the diverse pathways they are involved in. Therefore, the aim of this review is to provide an overview of the role of miRNAs in the progression and prognosis of gliomas, emphasizing the different pathways involved and identifying potential therapeutic targets.
Collapse
Affiliation(s)
- Omar Tluli
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Mazyona Al-Maadhadi
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Aisha Abdulla Al-Khulaifi
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Aishat F. Akomolafe
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Shaikha Y. Al-Kuwari
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | - Roudha Al-Khayarin
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| | | | - Shona Pedersen
- College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar; (O.T.); (M.A.-M.); (A.A.A.-K.); (A.F.A.); (R.A.-K.)
| |
Collapse
|
4
|
Hu Y, Zhang H, Guo Z, Zhou J, Zhang W, Gong M, Wu J. CKM and TERT dual promoters drive CRISPR-dCas9 to specifically inhibit the malignant behavior of osteosarcoma cells. Cell Mol Biol Lett 2023; 28:52. [PMID: 37415116 DOI: 10.1186/s11658-023-00464-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Improvements in treatment and chemotherapy have increased the survival rate of osteosarcoma, but overall efficacy remains low, highlighting the need for new gene therapy methods. Clustered regularly interspaced short palindromic repeats-deactivated Cas9 (CRISPR-dCas9) technology offers a promising strategy, but targeting osteosarcoma cells precisely is a challenge. We designed a system to achieve specific expression of CRISPR-dCas9-KRAB in osteosarcoma cells by using the creatine kinase muscle (CKM) promoter to drive dCas9-KRAB and the telomerase reverse transcriptase (TERT) promoter to drive single guide (sg)RNA expression. We inhibited the MDM2 proto-oncogene using this system in vitro, which efficiently inhibited the malignant behavior of osteosarcoma cells and induced apoptosis without affecting normal cells. In vivo experiments demonstrated that this system effectively inhibited the growth of subcutaneously transplanted tumors in nude mice. These findings provide a new method for precise identification and intervention of osteosarcoma with significant implications for the development of gene therapy methods for other cancers. Future research should focus on optimizing this system for clinical translation.
Collapse
Affiliation(s)
- Yawei Hu
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Hao Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Zengfeng Guo
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Jianhua Zhou
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Wang Zhang
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Ming Gong
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China
| | - Jiawen Wu
- Department of Spine Surgery, People's Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen, China.
| |
Collapse
|
5
|
Lin D, Li W, Zhang N, Cai M. Identification of TNFAIP6 as a hub gene associated with the progression of glioblastoma by weighted gene co-expression network analysis. IET Syst Biol 2022; 16:145-156. [PMID: 35766985 PMCID: PMC9469790 DOI: 10.1049/syb2.12046] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 05/29/2022] [Accepted: 06/08/2022] [Indexed: 11/19/2022] Open
Abstract
This study aims to discover the genetic modules that distinguish glioblastoma multiforme (GBM) from low‐grade glioma (LGG) and identify hub genes. A co‐expression network is constructed using the expression profiles of 28 GBM and LGG patients from the Gene Expression Omnibus database. The authors performed gene ontology (GO) and Kyoto encyclopaedia of genes and genomes (KEGG) analysis on these genes. The maximal clique centrality method was used to identify hub genes. Online tools were employed to confirm the link between hub gene expression and overall patient survival rate. The top 5000 genes with major variance were classified into 18 co‐expression gene modules. GO analysis indicated that abnormal changes in ‘cell migration’ and ‘collagen metabolic process’ were involved in the development of GBM. KEGG analysis suggested that ‘focal adhesion’ and ‘p53 signalling pathway’ regulate the tumour progression. TNFAIP6 was identified as a hub gene, and the expression of TNFAIP6 was increased with the elevation of pathological grade. Survival analysis indicated that the higher the expression of TNFAIP6, the shorter the survival time of patients. The authors identified TNFAIP6 as the hub gene in the progression of GBM, and its high expression indicates the poor prognosis of the patients.
Collapse
Affiliation(s)
- Dongdong Lin
- Department of Neurosurgery, The Second Affiliated Hospital-Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Li
- Department of Neurosurgery, The Second Affiliated Hospital-Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Nu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital-Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Cai
- Department of Neurosurgery, The Second Affiliated Hospital-Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
6
|
Grochans S, Cybulska AM, Simińska D, Korbecki J, Kojder K, Chlubek D, Baranowska-Bosiacka I. Epidemiology of Glioblastoma Multiforme-Literature Review. Cancers (Basel) 2022; 14:2412. [PMID: 35626018 PMCID: PMC9139611 DOI: 10.3390/cancers14102412] [Citation(s) in RCA: 183] [Impact Index Per Article: 91.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive malignancies, with a median overall survival of approximately 15 months. In this review, we analyze the pathogenesis of GBM, as well as epidemiological data, by age, gender, and tumor location. The data indicate that GBM is the higher-grade primary brain tumor and is significantly more common in men. The risk of being diagnosed with glioma increases with age, and median survival remains low, despite medical advances. In addition, it is difficult to determine clearly how GBM is influenced by stimulants, certain medications (e.g., NSAIDs), cell phone use, and exposure to heavy metals.
Collapse
Affiliation(s)
- Szymon Grochans
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich. 72 St., 70-111 Szczecin, Poland; (S.G.); (D.S.); (J.K.); (D.C.); (I.B.-B.)
| | - Anna Maria Cybulska
- Department of Nursing, Pomeranian Medical University in Szczecin, Żołnierska 48 St., 71-210 Szczecin, Poland
| | - Donata Simińska
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich. 72 St., 70-111 Szczecin, Poland; (S.G.); (D.S.); (J.K.); (D.C.); (I.B.-B.)
| | - Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich. 72 St., 70-111 Szczecin, Poland; (S.G.); (D.S.); (J.K.); (D.C.); (I.B.-B.)
- Department of Ruminants Science, Faculty of Biotechnology and Animal Husbandry, West Pomeranian University of Technology, Klemensa Janickiego 29 St., 71-270 Szczecin, Poland
| | - Klaudyna Kojder
- Department of Anaesthesiology and Intensive Care, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1 St., 71-281 Szczecin, Poland;
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich. 72 St., 70-111 Szczecin, Poland; (S.G.); (D.S.); (J.K.); (D.C.); (I.B.-B.)
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wielkopolskich. 72 St., 70-111 Szczecin, Poland; (S.G.); (D.S.); (J.K.); (D.C.); (I.B.-B.)
| |
Collapse
|
7
|
Rumex Vesicarius L. extract improves the efficacy of doxorubicin in triple-negative breast cancer through inhibiting Bcl2, mTOR, JNK1 and augmenting p21 expression. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100869] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
8
|
MicroRNAs Regulate Cell Cycle and Cell Death Pathways in Glioblastoma. Int J Mol Sci 2021; 22:ijms222413550. [PMID: 34948346 PMCID: PMC8705881 DOI: 10.3390/ijms222413550] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/25/2022] Open
Abstract
Glioblastoma (GBM), a grade IV brain tumor, is known for its heterogenicity and its resistance to the current treatment regimen. Over the last few decades, a significant amount of new molecular and genetic findings has been reported regarding factors contributing to GBM’s development into a lethal phenotype and its overall poor prognosis. MicroRNA (miRNAs) are small non-coding sequences of RNA that regulate and influence the expression of multiple genes. Many research findings have highlighted the importance of miRNAs in facilitating and controlling normal biological functions, including cell differentiation, proliferation, and apoptosis. Furthermore, miRNAs’ ability to initiate and promote cancer development, directly or indirectly, has been shown in many types of cancer. There is a clear association between alteration in miRNAs expression in GBM’s ability to escape apoptosis, proliferation, and resistance to treatment. Further, miRNAs regulate the already altered pathways in GBM, including P53, RB, and PI3K-AKT pathways. Furthermore, miRNAs also contribute to autophagy at multiple stages. In this review, we summarize the functions of miRNAs in GBM pathways linked to dysregulation of cell cycle control, apoptosis and resistance to treatment, and the possible use of miRNAs in clinical settings as treatment and prediction biomarkers.
Collapse
|
9
|
Farzaneh Z, Vosough M, Agarwal T, Farzaneh M. Critical signaling pathways governing hepatocellular carcinoma behavior; small molecule-based approaches. Cancer Cell Int 2021; 21:208. [PMID: 33849569 PMCID: PMC8045321 DOI: 10.1186/s12935-021-01924-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/07/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of death due to cancer. Although there are different treatment options, these strategies are not efficient in terms of restricting the tumor cell's proliferation and metastasis. The liver tumor microenvironment contains the non-parenchymal cells with supportive or inhibitory effects on the cancerous phenotype of HCC. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of liver carcinoma cells. Recent studies have established new approaches for the prevention and treatment of HCC using small molecules. Small molecules are compounds with a low molecular weight that usually inhibit the specific targets in signal transduction pathways. These components can induce cell cycle arrest, apoptosis, block metastasis, and tumor growth. Devising strategies for simultaneously targeting HCC and the non-parenchymal population of the tumor could lead to more relevant research outcomes. These strategies may open new avenues for the treatment of HCC with minimal cytotoxic effects on healthy cells. This study provides the latest findings on critical signaling pathways governing HCC behavior and using small molecules in the control of HCC both in vitro and in vivo models.
Collapse
Affiliation(s)
- Zahra Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, 721302, India
| | - Maryam Farzaneh
- Fertility, Infertility and Perinatology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
10
|
Banerjee K, Núñez FJ, Haase S, McClellan BL, Faisal SM, Carney SV, Yu J, Alghamri MS, Asad AS, Candia AJN, Varela ML, Candolfi M, Lowenstein PR, Castro MG. Current Approaches for Glioma Gene Therapy and Virotherapy. Front Mol Neurosci 2021; 14:621831. [PMID: 33790740 PMCID: PMC8006286 DOI: 10.3389/fnmol.2021.621831] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in the adult population and it carries a dismal prognosis. Inefficient drug delivery across the blood brain barrier (BBB), an immunosuppressive tumor microenvironment (TME) and development of drug resistance are key barriers to successful glioma treatment. Since gliomas occur through sequential acquisition of genetic alterations, gene therapy, which enables to modification of the genetic make-up of target cells, appears to be a promising approach to overcome the obstacles encountered by current therapeutic strategies. Gene therapy is a rapidly evolving field with the ultimate goal of achieving specific delivery of therapeutic molecules using either viral or non-viral delivery vehicles. Gene therapy can also be used to enhance immune responses to tumor antigens, reprogram the TME aiming at blocking glioma-mediated immunosuppression and normalize angiogenesis. Nano-particles-mediated gene therapy is currently being developed to overcome the BBB for glioma treatment. Another approach to enhance the anti-glioma efficacy is the implementation of viro-immunotherapy using oncolytic viruses, which are immunogenic. Oncolytic viruses kill tumor cells due to cancer cell-specific viral replication, and can also initiate an anti-tumor immunity. However, concerns still remain related to off target effects, and therapeutic and transduction efficiency. In this review, we describe the rationale and strategies as well as advantages and disadvantages of current gene therapy approaches against gliomas in clinical and preclinical studies. This includes different delivery systems comprising of viral, and non-viral delivery platforms along with suicide/prodrug, oncolytic, cytokine, and tumor suppressor-mediated gene therapy approaches. In addition, advances in glioma treatment through BBB-disruptive gene therapy and anti-EGFRvIII/VEGFR gene therapy are also discussed. Finally, we discuss the results of gene therapy-mediated human clinical trials for gliomas. In summary, we highlight the progress, prospects and remaining challenges of gene therapies aiming at broadening our understanding and highlighting the therapeutic arsenal for GBM.
Collapse
Affiliation(s)
- Kaushik Banerjee
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Felipe J. Núñez
- Laboratory of Molecular and Cellular Therapy, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Brandon L. McClellan
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Immunology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Syed M. Faisal
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephen V. Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Cancer Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Jin Yu
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Mahmoud S. Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Antonela S. Asad
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro J. Nicola Candia
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Maria Luisa Varela
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Marianela Candolfi
- Departamento de Biología e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedro R. Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
11
|
Ibnat N, Kamaruzman NI, Ashaie M, Chowdhury EH. Transfection with p21 and p53 tumor suppressor plasmids suppressed breast tumor growth in syngeneic mouse model. Gene 2019; 701:32-40. [PMID: 30898703 DOI: 10.1016/j.gene.2019.02.082] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 02/16/2019] [Accepted: 02/23/2019] [Indexed: 01/13/2023]
Abstract
Treatment of breast cancer by delivering important tumor suppressor plasmids is a promising approach in the field of clinical medicine. We transfected p21 and p53 tumor suppressor plasmids, into different breast cancer cell lines using inorganic nanoparticles (NPs) of carbonate apatite to evaluate the effect of gene expression on reducing breast cancer cell growth. In triple negative MDA-MB-231 breast cancer cell line, the cytotoxicity assay upon combined delivery of p21 and p53 plasmid loaded NPs showed significant decrease in cell growth compared to distinct p21 or p53 treatments. Also, in MCF-7 and 4T1 cell lines, significant reduction in cellular growth was observed following p21 or p53 plasmid transfection. The Western blot data showed that NP loaded p21 and p53 transgene delivery in MDA-MB-231 cell line resulted in a noteworthy decrease in phosphorylated form of MAPK protein of MAPK/ERK pathway. The in vivo studies in syngeneic breast cancer mouse model demonstrated that the rate of growth and final tumor volume were reduced to a greater extent in mice that received intravenous injection of p21 + NP and p53 + NP therapeutics.
Collapse
Affiliation(s)
- Nabilah Ibnat
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | | | - Maeirah Ashaie
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Australia
| | | |
Collapse
|
12
|
Lee JY, Lee SH, Kim KS, Park KH, Park KS. Ell3 functions as a critical decision maker at the crossroad between stem cell senescence and apoptosis. Stem Cell Res Ther 2019; 10:32. [PMID: 30654843 PMCID: PMC6335702 DOI: 10.1186/s13287-019-1137-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 12/16/2018] [Accepted: 01/07/2019] [Indexed: 11/23/2022] Open
Abstract
Background Ell3 is a RNA polymerase II elongation factor that has various cell type-dependent functions, such as regulating the differentiation efficiency of embryonic stem cells and sensitizing cancer cells to anticancer drugs. However, there has been little research on the role of Ell3 on the regulation of senescence and apoptosis of stem cells. Methods We analyzed the senescence of Ell3-suppressed stem cells by mitochondrial activity, β-gal (+) cells, and lineage differentiation efficiency. The apoptosis of Ell3-overexpressing stem cells was analyzed by Annexin V staining, Immunoblot, and Live&dead assay. In addition, chromatin immunoprecipitation and luciferase assays were used to demonstrate p53 functions as a direct transcriptional activator of Ell3. Results Suppression of Ell3 expression induced senescence in stem cells by increasing Bcl-2 expression. Unlike the effect of Ell3 suppression, the ectopic expression of Ell3 induces apoptosis of stem cells and induces apoptosis of adjacent cells. In addition, p53 functions as a direct transcriptional activator of Ell3 during the stem cell apoptosis. Conclusions We suggest that the function of Ell3 is associated with the p53-Bcl2 axis in both senescent and apoptotic ADSCs. Electronic supplementary material The online version of this article (10.1186/s13287-019-1137-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jae-Yong Lee
- Department of Biomedical Science, College of Life Science and CHA Stem Cell Institute, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, Korea
| | - Soo-Hong Lee
- Department of Biomedical Science, College of Life Science and CHA Stem Cell Institute, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, Korea
| | - Kwang-Soo Kim
- Department of Biomedical Science, College of Life Science and CHA Stem Cell Institute, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, Korea
| | - Keun-Hong Park
- Department of Biomedical Science, College of Life Science and CHA Stem Cell Institute, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, Korea
| | - Kyung-Soon Park
- Department of Biomedical Science, College of Life Science and CHA Stem Cell Institute, CHA University, 335, Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-400, Korea.
| |
Collapse
|
13
|
The p53 Pathway in Glioblastoma. Cancers (Basel) 2018; 10:cancers10090297. [PMID: 30200436 PMCID: PMC6162501 DOI: 10.3390/cancers10090297] [Citation(s) in RCA: 227] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 12/27/2022] Open
Abstract
The tumor suppressor and transcription factor p53 plays critical roles in tumor prevention by orchestrating a wide variety of cellular responses, including damaged cell apoptosis, maintenance of genomic stability, inhibition of angiogenesis, and regulation of cell metabolism and tumor microenvironment. TP53 is one of the most commonly deregulated genes in cancer. The p53-ARF-MDM2 pathway is deregulated in 84% of glioblastoma (GBM) patients and 94% of GBM cell lines. Deregulated p53 pathway components have been implicated in GBM cell invasion, migration, proliferation, evasion of apoptosis, and cancer cell stemness. These pathway components are also regulated by various microRNAs and long non-coding RNAs. TP53 mutations in GBM are mostly point mutations that lead to a high expression of a gain of function (GOF) oncogenic variants of the p53 protein. These relatively understudied GOF p53 mutants promote GBM malignancy, possibly by acting as transcription factors on a set of genes other than those regulated by wild type p53. Their expression correlates with worse prognosis, highlighting their potential importance as markers and targets for GBM therapy. Understanding mutant p53 functions led to the development of novel approaches to restore p53 activity or promote mutant p53 degradation for future GBM therapies.
Collapse
|
14
|
Adjiri A. Tracing the path of cancer initiation: the AA protein-based model for cancer genesis. BMC Cancer 2018; 18:831. [PMID: 30119662 PMCID: PMC6098654 DOI: 10.1186/s12885-018-4739-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/09/2018] [Indexed: 02/07/2023] Open
Abstract
Background Cancer is a defiant disease which cure is still far from being attained besides the colossal efforts and financial means deployed towards that end. The continuing setbacks encountered with today’s arsenal of anti-cancer drugs and cancer therapy modalities; show the need for a radical approach in order to get to the root of the problem. And getting to the root of cancer initiation and development leads us to challenge the present dogmas surrounding the pathogenesis of this disease. Results This comprehensive analysis brings to light the following points: (i) Cancer with its plethora of genetic and cellular symptoms could originate from one major event switching a cell from normalcy-to-malignancy; (ii) The switching event is postulated to involve a pathological breakup of a non-mutated protein, called here AA protein, resulting in the acquisition of new cellular functions present only in cancer cells; (iii) Following this event, DNA mutations begin to accumulate as secondary events to ensure perpetuity of cancer. Supporting arguments for this protein-based model come mainly from these observations: (i) The AA protein-based model reconciles together the clonal-and-stem cell theories into one inclusive model; (ii) The breakup of a normal protein could be behind the cancer-linked inflammation symptom; (iii) Cancer hallmarks are but adaptive traits, earned as a result of the switch from normalcy-to-malignancy. Conclusions Adaptation of cancer cells to their microenvironment and to different anti-cancer drugs is deemed here as the ultimate cancer hallmark, that needs to be understood and controlled. This adaptive power of cancer cells parallels that of bacteria also known with their resistance to a large range of substances in nature and in the laboratory. Consequently, cancer development could be viewed as a backward walk on the line of Evolution. Finally this unprecedented analysis demystifies cancer and puts the finger on the core problem of malignancy while offering ideas for its control with the ultimate goal of leading to its cure.
Collapse
Affiliation(s)
- Adouda Adjiri
- Physics Department, Faculty of Sciences, Sétif-1 University, 19000, Sétif, Algeria.
| |
Collapse
|
15
|
Mangraviti A, Tzeng SY, Kozielski KL, Wang Y, Jin Y, Gullotti D, Pedone M, Buaron N, Liu A, Wilson DR, Hansen SK, Rodriguez FJ, Gao GD, DiMeco F, Brem H, Olivi A, Tyler B, Green JJ. Polymeric nanoparticles for nonviral gene therapy extend brain tumor survival in vivo. ACS NANO 2015; 9:1236-49. [PMID: 25643235 PMCID: PMC4342728 DOI: 10.1021/nn504905q] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Biodegradable polymeric nanoparticles have the potential to be safer alternatives to viruses for gene delivery; however, their use has been limited by poor efficacy in vivo. In this work, we synthesize and characterize polymeric gene delivery nanoparticles and evaluate their efficacy for DNA delivery of herpes simplex virus type I thymidine kinase (HSVtk) combined with the prodrug ganciclovir (GCV) in a malignant glioma model. We investigated polymer structure for gene delivery in two rat glioma cell lines, 9L and F98, to discover nanoparticle formulations more effective than the leading commercial reagent Lipofectamine 2000. The lead polymer structure, poly(1,4-butanediol diacrylate-co-4-amino-1-butanol) end-modified with 1-(3-aminopropyl)-4-methylpiperazine, is a poly(β-amino ester) (PBAE) and formed nanoparticles with HSVtk DNA that were 138 ± 4 nm in size and 13 ± 1 mV in zeta potential. These nanoparticles containing HSVtk DNA showed 100% cancer cell killing in vitro in the two glioma cell lines when combined with GCV exposure, while control nanoparticles encoding GFP maintained robust cell viability. For in vivo evaluation, tumor-bearing rats were treated with PBAE/HSVtk infusion via convection-enhanced delivery (CED) in combination with systemic administration of GCV. These treated animals showed a significant benefit in survival (p = 0.0012 vs control). Moreover, following a single CED infusion, labeled PBAE nanoparticles spread completely throughout the tumor. This study highlights a nanomedicine approach that is highly promising for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Stephany Yi Tzeng
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Kristen Lynn Kozielski
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Yuan Wang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Yike Jin
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - David Gullotti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Mariangela Pedone
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Nitsa Buaron
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Chemical Engineering, Ben Gurion University of the Negev, Be’er Sheva 84105, Israel
| | - Ann Liu
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - David R. Wilson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Sarah K. Hansen
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Fausto J. Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi’an 710032, China
| | - Francesco DiMeco
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico C. Besta, Milan 20133, Italy
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| | - Jordan J. Green
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- The Institute for Nanobiotechnology and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21231, United States
- Department of Material Science and Engineering, Johns Hopkins University, Baltimore, Maryland 21231, United States
- Address correspondence to ,
| |
Collapse
|
16
|
Damdindorj L, Karnan S, Ota A, Hossain E, Konishi Y, Hosokawa Y, Konishi H. A comparative analysis of constitutive promoters located in adeno-associated viral vectors. PLoS One 2014; 9:e106472. [PMID: 25170953 PMCID: PMC4149579 DOI: 10.1371/journal.pone.0106472] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 08/07/2014] [Indexed: 11/18/2022] Open
Abstract
The properties of constitutive promoters within adeno-associated viral (AAV) vectors have not yet been fully characterized. In this study, AAV vectors, in which enhanced GFP expression was directed by one of the six constitutive promoters (human β-actin, human elongation factor-1α, chicken β-actin combined with cytomegalovirus early enhancer, cytomegalovirus (CMV), simian virus 40, and herpes simplex virus thymidine kinase), were constructed and introduced into the HCT116, DLD-1, HT-1080, and MCF-10A cell lines. Quantification of GFP signals in infected cells demonstrated that the CMV promoter produced the highest GFP expression in the six promoters and maintained relatively high GFP expression for up to eight weeks after infection of HCT116, DLD-1, and HT-1080. Exogenous human CDKN2A gene expression was also introduced into DLD-1 and MCF-10A in a similar pattern by using AAV vectors bearing the human β-actin and the CMV promoters. The six constitutive promoters were subsequently placed upstream of the neomycin resistance gene within AAV vectors, and HCT116, DLD-1, and HT-1080 were infected with the resulting vectors. Of the six promoters, the CMV promoter produced the largest number of G418-resistant colonies in all three cell lines. Because AAV vectors have been frequently used as a platform to construct targeting vectors that permit gene editing in human cell lines, we lastly infected the three cell lines with AAV-based targeting vectors against the human PIGA gene in which one of the six promoters regulate the neomycin resistance gene. This assay revealed that the CMV promoter led to the lowest PIGA gene targeting efficiency in the investigated promoters. These results provide a clue to the identification of constitutive promoters suitable to express exogenous genes with AAV vectors, as well as those helpful to conduct efficient gene targeting using AAV-based targeting vectors in human cell lines.
Collapse
Affiliation(s)
- Lkhagvasuren Damdindorj
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Ekhtear Hossain
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Yuko Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine Nagakute, Aichi, Japan
- * E-mail:
| |
Collapse
|
17
|
Chen S, Zhao D, Li F, Zhuo RX, Cheng SX. Co-delivery of genes and drugs with nanostructured calcium carbonate for cancer therapy. RSC Adv 2012. [DOI: 10.1039/c1ra00527h] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
18
|
Castro MG, Candolfi M, Kroeger K, King GD, Curtin JF, Yagiz K, Mineharu Y, Assi H, Wibowo M, Ghulam Muhammad AKM, Foulad D, Puntel M, Lowenstein PR. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2011; 11:155-80. [PMID: 21453286 DOI: 10.2174/156652311795684722] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of 15-18 months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted; this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Maria G Castro
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
van Putten EH, Dirven CM, van den Bent MJ, Lamfers ML. Sitimagene ceradenovec: a gene-based drug for the treatment of operable high-grade glioma. Future Oncol 2011; 6:1691-710. [PMID: 21142657 DOI: 10.2217/fon.10.134] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The field of gene therapy for malignant glioma has made important advances since the first gene transfer studies were performed 20 years ago. Multiple Phase I/II trials and two Phase III trials have been performed and have demonstrated the feasibility and safety of intratumoral vector delivery in the brain. Sitimagene ceradenovec is an adenoviral vector encoding the herpes simplex thymidine kinase gene, developed by Ark Therapeutics Group plc (UK and Finland) for the treatment of patients with operable high-grade glioma. In preclinical and Phase I/II clinical studies, sitimagene ceradenovec exhibited a significant increase in survival. Although the preliminary results of a Phase III clinical study demonstrated a significant positive effect of sitimagene ceradenovec treatment on time to reintervention or death when compared with standard care treatment (hazard ratio: 1.43; 95% CI: 1.06-1.93; p < 0.05), the European Committee for Medicinal Products for Human Use did not consider the data to provide sufficient evidence of clinical benefit. Further clinical evaluation, powered to demonstrate a benefit on a robust end point, is required. This article focuses on sitimagene ceradenovec and provides an overview of the developments in the field of gene therapy for malignant glioma.
Collapse
Affiliation(s)
- Erik Hp van Putten
- Department of Neurosurgery, Erasmus University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands.
| | | | | | | |
Collapse
|
20
|
Sun W, Yang J. Functional mechanisms for human tumor suppressors. J Cancer 2010; 1:136-40. [PMID: 20922055 PMCID: PMC2948218 DOI: 10.7150/jca.1.136] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 09/13/2010] [Indexed: 12/19/2022] Open
Abstract
Tumor suppressors refer to a large group of molecules that are capable of controlling cell division, promoting apoptosis, and suppressing metastasis. The loss of function for a tumor suppressor may lead to cancer due to uncontrolled cell division. Because of their importance, extensive studies have been undertaken to understand the different functional mechanisms of tumor suppressors. Here, we briefly review the four major mechanisms, inhibition of cell division, induction of apoptosis, DNA damage repair, and inhibition of metastasis. It is noteworthy that some tumor suppressors, such as p53, may adopt more than one mechanism for their functions.
Collapse
Affiliation(s)
- Wanpeng Sun
- Drug Design and Discovery Research Group, College of Pharmacy and Nutrition, University of Saskatchewan, 110 Science Place, Saskatoon, Saskatchewan S7N 5C9, Canada
| | | |
Collapse
|
21
|
Bai J, Meng Z. Effect of sulfur dioxide on expression of proto-oncogenes and tumor suppressor genes from rats. ENVIRONMENTAL TOXICOLOGY 2010; 25:272-283. [PMID: 19434695 DOI: 10.1002/tox.20495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Sulfur dioxide (SO(2)) is a ubiquitous air pollutant that is present in low concentrations in the urban air, and in higher concentrations in the working environment. In the present study, male Wistar rats were housed in exposure chambers and treated with 14.00 +/- 1.01, 28.00 +/- 1.77 and 56.00 +/- 3.44 mg m(-3) SO(2) for 6 h/day for 7 days, while control group was exposed to filtered air in the same condition. The mRNA and protein levels of proto-oncogenes (c-fos, c-jun, c-myc, and Ki-ras) and tumor suppressor genes (p53, Rb, and p16) were analyzed in lungs using a real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) assay and Western blot analysis. The results showed that mRNA and protein levels of c-fos, c-jun, c-myc, Ki-ras, and p53 in lungs were increased in a dose-dependent manner, while mRNA and protein levels of Rb and p16 were decreased in lungs of rats after SO(2) inhalation. These results lead to a conclusion that SO(2) exposure could activate expressions of proto-oncogenes and suppress expressions of tumor suppressor genes, which might relate to the molecular mechanism of cocarcinogenic properties and potential carcinogenic effects of SO(2). According to previous studies, the results also indicated that promoter genes of apoptosis and tumor suppressor genes could produce apoptotic signals to antagonize the growth signals that arise from oncogenes. Understanding its molecular controls will benefit development of treatments for many diseases.
Collapse
Affiliation(s)
- Juli Bai
- Institute of Environmental Medicine and Toxicology, Research Center of Environmental Science and Engineering, Shanxi University, Taiyuan, China
| | | |
Collapse
|
22
|
Song T, Wu J, Fang F, Chen F, Huo L, Zhang M, Wu L, Zhai Z, Yang L, Fang J. Correlation analysis between the expression of P21WAF1/CIP1, P16 proteins and human glioma. Clin Exp Med 2008; 8:151-7. [PMID: 18791688 DOI: 10.1007/s10238-008-0172-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 02/26/2008] [Indexed: 11/25/2022]
Abstract
AIM Glioma is the most common neoplasm of the brain. Unfortunately, surgical cure of it is practically impossible and clinical course is primarily determined by the biological behaviour of the tumour cells. The purpose of this study was to investigate the correlation between the expression levels of P21WAF1/CIP1, P16 proteins and the grading of glioma. METHODS T98G human glioma cell line, normal human astrocyte (HA) cell line, tumour tissue samples from 70 patients suffering from glioma and normal brain tissues from 20 cases with brain contusion were investigated. The expression levels of P21WAF1/CIP1 and P16 proteins were detected using SABC immunohistochemical staining and semi-quantitive reverse transcriptase polymerase chain reaction (RT-PCR) assay. Then, the correlation of the two markers' expression with glioma grading of patients was analysed. RESULTS The expression levels of P21WAF1/CIP1 and P16 proteins in the T98G cell line were much lower than that in the HA cell line. Their positive expression rates in glioma tissues were 55.71% and 42.86% respectively, and a significant increase was observed in normal brain tissues (p = 0.012, 0.008). Combined with the result of semi-quantitive RT-PCR, we could demonstrate that the expression intensity of P21WAF1/CIP1 and P16 decreased with the glioma grade increase. Co-expression of them was also found in glioma and normal brain tissues. Furthermore, there was a negative correlation between the two markers' expression and glioma grading of patients (rs = -0.68, -0.56). CONCLUSIONS The positive expression rate and co-expression rate of P21WAF1/CIP1 and P16 proteins could reflect the malignant grade of glioma to some extent, and they can be considered as a sensitive index for glioma grading.
Collapse
Affiliation(s)
- Tao Song
- Neurosurgery Department, Xiangya Hospital of Central South University, Changsha, Hunan, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Organotin(IV) complexes of 2-pyridineformamide-derived thiosemicarbazones: Antimicrobial and cytotoxic effects. Eur J Med Chem 2008; 43:1454-61. [PMID: 17983689 DOI: 10.1016/j.ejmech.2007.09.016] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Revised: 09/18/2007] [Accepted: 09/19/2007] [Indexed: 11/23/2022]
|
24
|
King GD, Curtin JF, Candolfi M, Kroeger K, Lowenstein PR, Castro MG. Gene therapy and targeted toxins for glioma. Curr Gene Ther 2006; 5:535-57. [PMID: 16457645 PMCID: PMC1629033 DOI: 10.2174/156652305774964631] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The most common primary brain tumor in adults is glioblastoma. These tumors are highly invasive and aggressive with a mean survival time of nine to twelve months from diagnosis to death. Current treatment modalities are unable to significantly prolong survival in patients diagnosed with glioblastoma. As such, glioma is an attractive target for developing novel therapeutic approaches utilizing gene therapy. This review will examine the available preclinical models for glioma including xenographs, syngeneic and genetic models. Several promising therapeutic targets are currently being pursued in pre-clinical investigations. These targets will be reviewed by mechanism of action, i.e., conditional cytotoxic, targeted toxins, oncolytic viruses, tumor suppressors/oncogenes, and immune stimulatory approaches. Preclinical gene therapy paradigms aim to determine which strategies will provide rapid tumor regression and long-term protection from recurrence. While a wide range of potential targets are being investigated preclinically, only the most efficacious are further transitioned into clinical trial paradigms. Clinical trials reported to date are summarized including results from conditionally cytotoxic, targeted toxins, oncolytic viruses and oncogene targeting approaches. Clinical trial results have not been as robust as preclinical models predicted, this could be due to the limitations of the GBM models employed. Once this is addressed, and we develop effective gene therapies in models that better replicate the clinical scenario, gene therapy will provide a powerful approach to treat and manage brain tumors.
Collapse
Affiliation(s)
- Gwendalyn D King
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | | | | | | | | |
Collapse
|
25
|
Ranuncolo SM, Varela M, Morandi A, Lastiri J, Christiansen S, Bal de Kier Joffé E, Pallotta MG, Puricelli L. Prognostic value of Mdm2, p53 and p16 in patients with astrocytomas. J Neurooncol 2004; 68:113-21. [PMID: 15218947 DOI: 10.1023/b:neon.0000027741.19213.99] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Surgical cure of gliomas infiltrating into the brain is practically impossible and their clinical course is primarily determined by the biological behavior of the tumor cell. The purpose of this study was to analyze retrospectively prognostic input of p53, Mouse double minute-2 (Mdm2) and p16 in 103 uniformly treated patients with astrocytic tumors. The expression of these molecules was measured by immunohistochemical procedure. Prognostic evaluation was performed with the multivariate proportional hazards model. The follow-up period lasted 19 (5-122) months for the survivors. We observed that 66% of gliomas showed mutated p53, while only 17% overexpressed Mdm2, the p53-regulatory molecule. Besides, almost 50% of gliomas lost p16 immunopositivity. Only p53 labeling showed a positive correlation with the grade of malignancy, according with the WHO classification. The association between mutated p53 and histological grade remained when prognostic variables were considered in a multivariate analysis. No association between p53 status and overall survival was found. On the other hand, Mdm2 overexpression and, unexpectedly, p16 immunopositivity were associated with a shorter survival in an univariate analysis. However, Cox-regression analysis showed that only Mdm2 in female patients was an independent prognostic factor, associated with shorter survival. In conclusion, our results suggest that Mdm2 could be a relevant marker in determining the evolution of glioma patients and could provide a more objective way to classify astrocytomas.
Collapse
Affiliation(s)
- Stella M Ranuncolo
- Research Area of the Institute of Oncology Angel H. Roffo, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Kanzawa T, Ito H, Kondo Y, Kondo S. Current and Future Gene Therapy for Malignant Gliomas. J Biomed Biotechnol 2003; 2003:25-34. [PMID: 12686720 PMCID: PMC179758 DOI: 10.1155/s1110724303209013] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Malignant gliomas are the most common neoplasm in the central nervous system. When treated with conventional treatments including surgery, irradiation, and chemotherapy, the average life expectancy of the most malignant type, glioblastoma multiforme is usually less than 1 year. Therefore, gene therapy is expected to be an effective and possibly curative treatment. Many gene therapeutic approaches have demonstrated efficacy in experimental animal models. However, the current clinical trials are disappointing. This review focuses on current therapeutic genes/vectors/delivery systems/targeting strategies in order to introduce updated trends and hopefully indicate prospective gene therapy for malignant gliomas.
Collapse
Affiliation(s)
- Takao Kanzawa
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Hideaki Ito
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasuko Kondo
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Seiji Kondo
- Department of Neurosurgery, Mount Sinai School of Medicine, New York, NY 10029, USA
- Department of Neurosurgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
27
|
Huang JZ, Xia SS, Ye QF, Jiang HY, Chen ZH. Effects of p16 gene on biological behavious in hepatocellular carcimoma cells. World J Gastroenterol 2003; 9:84-8. [PMID: 12508357 PMCID: PMC4728255 DOI: 10.3748/wjg.v9.i1.84] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of p16 gene on biological behavious in hepatocellular carcinoma cells.
METHODS: HCC cell lines SNU-449 and HepG2.2.15 were infected respectively by a replication defective, recombinant retrovirus capable of producing a high level of p16 protein expression (pCLXSN-p16). G418 resistant stable p16 protein expression cell lines were selected. And the biological behaviours of the p16 gene transfected HCC cells were observed.
RESULTS: Initial in vitro experiments in HCC cell line SNU-449 with loss of p16 protein expression demonstrated the pCLXSN-p16 treatment significantly inhibited cell growth. But there was no treatment effect when the pCLXSN-p16 was used in another HCC cell line HepG2.2.15 which has positive p16 protein expression. Subsequent study in a nude mouse model demonstrated that the p16 gene transfected SNU-449 had a lower succeeding rate in the first time establishment of tumors and grew more slowly in the nude mice when compared with non-transfected SNU-449. Moreover, the nude mice inoculated with transfected SNU-449 had a longer surviving time than those inoculated with non-transfected SNU-449.
CONCLUSION: Our results show that the p16INK4a gene transfer can inhibit the proliferation and reduce the invasion ability of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Jian-Zhao Huang
- Department of Hepatobilliary Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | | | | | | | | |
Collapse
|