1
|
Miles X, Vandevoorde C, Hunter A, Bolcaen J. MDM2/X Inhibitors as Radiosensitizers for Glioblastoma Targeted Therapy. Front Oncol 2021; 11:703442. [PMID: 34307171 PMCID: PMC8296304 DOI: 10.3389/fonc.2021.703442] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Inhibition of the MDM2/X-p53 interaction is recognized as a potential anti-cancer strategy, including the treatment of glioblastoma (GB). In response to cellular stressors, such as DNA damage, the tumor suppression protein p53 is activated and responds by mediating cellular damage through DNA repair, cell cycle arrest and apoptosis. Hence, p53 activation plays a central role in cell survival and the effectiveness of cancer therapies. Alterations and reduced activity of p53 occur in 25-30% of primary GB tumors, but this number increases drastically to 60-70% in secondary GB. As a result, reactivating p53 is suggested as a treatment strategy, either by using targeted molecules to convert the mutant p53 back to its wild type form or by using MDM2 and MDMX (also known as MDM4) inhibitors. MDM2 down regulates p53 activity via ubiquitin-dependent degradation and is amplified or overexpressed in 14% of GB cases. Thus, suppression of MDM2 offers an opportunity for urgently needed new therapeutic interventions for GB. Numerous small molecule MDM2 inhibitors are currently undergoing clinical evaluation, either as monotherapy or in combination with chemotherapy and/or other targeted agents. In addition, considering the major role of both p53 and MDM2 in the downstream signaling response to radiation-induced DNA damage, the combination of MDM2 inhibitors with radiation may offer a valuable therapeutic radiosensitizing approach for GB therapy. This review covers the role of MDM2/X in cancer and more specifically in GB, followed by the rationale for the potential radiosensitizing effect of MDM2 inhibition. Finally, the current status of MDM2/X inhibition and p53 activation for the treatment of GB is given.
Collapse
Affiliation(s)
- Xanthene Miles
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Alistair Hunter
- Radiobiology Section, Division of Radiation Oncology, Department of Radiation Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| |
Collapse
|
2
|
Dougherty PG, Wen J, Pan X, Koley A, Ren JG, Sahni A, Basu R, Salim H, Appiah Kubi G, Qian Z, Pei D. Enhancing the Cell Permeability of Stapled Peptides with a Cyclic Cell-Penetrating Peptide. J Med Chem 2019; 62:10098-10107. [PMID: 31657556 DOI: 10.1021/acs.jmedchem.9b00456] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stapled peptides recapitulate the binding affinity and specificity of α-helices in proteins, resist proteolytic degradation, and may provide a novel modality against challenging drug targets such as protein-protein interactions. However, most of the stapled peptides have limited cell permeability or are impermeable to the cell membrane. We show herein that stapled peptides can be rendered highly cell-permeable by conjugating a cyclic cell-penetrating peptide to their N-terminus, C-terminus, or stapling unit. Application of this strategy to two previously reported membrane-impermeable peptidyl inhibitors against the MDM2/p53 and β-catenin/TCF interactions resulted in the generation of potent proof-of-concept antiproliferative agents against key therapeutic targets.
Collapse
Affiliation(s)
- Patrick G Dougherty
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States.,Entrada Therapeutics Inc. , 50 Northern Avenue , Boston , Massachusetts 02210 , United States
| | - Jin Wen
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Xiaoyan Pan
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Amritendu Koley
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Jian-Guo Ren
- Entrada Therapeutics Inc. , 50 Northern Avenue , Boston , Massachusetts 02210 , United States
| | - Ashweta Sahni
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Ruchira Basu
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Heba Salim
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - George Appiah Kubi
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| | - Ziqing Qian
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States.,Entrada Therapeutics Inc. , 50 Northern Avenue , Boston , Massachusetts 02210 , United States
| | - Dehua Pei
- Department of Chemistry and Biochemistry , The Ohio State University , Columbus , Ohio 43210 , United States
| |
Collapse
|
3
|
Wang D, Guo M, Yu J, Wang X, Zhang Q, Yang X, Li J, Zhao C, Feng B. Glioma targeting peptide in combination with the P53 C terminus inhibits glioma cell proliferation in vitro. Cytotechnology 2017; 70:153-161. [PMID: 28879517 DOI: 10.1007/s10616-017-0122-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 07/07/2017] [Indexed: 10/18/2022] Open
Abstract
Glioma is a prevalent malignant primary brain tumor in adults, the treatment for which remains a challenge due to its high infiltration and recurrence. Hence, treatments that lead to the suppression of glioma cell migration and invasion may be used in addition to surgery to increase the therapeutic outcome. In this study, we aimed to construct a multifunctional protein that would exert an effect on glioma cell proliferation and migration. The protein is named GL1-P53C-11R and it consists of the glioma-targeting peptide GL1 (G), the P53 C terminus (Pc) and the cell-penetrating peptide arginine (R). GL1-P53C-R was expressed with the fusion protein ZZ and immunofluorescence analysis showed effective delivery of the fused ZZ-GL1-P53C-R protein represented as ZZ-GPcR. The ZZ-GPcR exhibited an inhibitory effect on the proliferation, migration and invasion of U87ΔEGFR cells. Western blotting results indicated that it caused significant changes in the expression levels of cell cycle and apoptotic proteins. Flow cytometric analysis showed increase apoptosis. Our findings suggest that the P53C in the fusion protein ZZ-GPcR can enter into glioma cells to exert its inhibitory effect.
Collapse
Affiliation(s)
- Dan Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Meihua Guo
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Jiawen Yu
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xinying Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Qian Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xu Yang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Jiaqi Li
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Chunhui Zhao
- College of Life Sciences, Liaoning Normal University, Dalian, 116029, China.
| | - Bin Feng
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
4
|
Lu J, Lin L, Dong H, Meng X, Fang F, Wang Q, Huang L, Tan J. Protein therapy using MafA fused to a polyarginine transduction domain attenuates glucose levels of streptozotocin‑induced diabetic mice. Mol Med Rep 2017; 15:4041-4048. [PMID: 28487936 PMCID: PMC5436157 DOI: 10.3892/mmr.2017.6536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 02/21/2017] [Indexed: 12/02/2022] Open
Abstract
Ectopic expression of musculo aponeurotic fibrosarcoma BZIP transcription factor (Maf) A, has previously been demonstrated to induce insulin expression in non-β-cell lines. Protein transduction domains acting as an alternative delivery strategy may deliver heterogeneous proteins into cells. A sequence of 11 arginine residues (11R) has been demonstrated to act as a particularly efficient vector to introduce proteins into various cell types. The present study constructed 11R-fused MafA to achieve transduction of the protein into cellular membranes and subsequently examined the therapeutic effect of the MafA-11R protein in streptozotocin-induced diabetes. A small animal imaging system was used to demonstrate that 11R introduced proteins into cells. The MafA-11R protein was then injected into the tale vein of healthy male mice, and western blot analysis and immunofluorescence staining was performed to identify the location of the recombinant protein. Ameliorated hyperglycemia in the MafA-11R-treated diabetic mice was demonstrated via the improved intraperitoneal glucose tolerance test (IPGTT) and glucose-stimulated insulin release. Furthermore, insulin producing cells were detected in the jejunum of the MafA-11R treated mice. The results of the present study indicated that MafA-11R delivery may act as a novel and potential therapeutic strategy for the future and will not present adverse effects associated with viral vector-mediated gene therapies.
Collapse
Affiliation(s)
- Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Lingjing Lin
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Huiyue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Xin Meng
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Fang Fang
- Department of Stomatology, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Qinghua Wang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Lianghu Huang
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Jianming Tan
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
5
|
Wang N, Sun P, Lv M, Tong G, Jin X, Zhu X. Mustard-inspired delivery shuttle for enhanced blood–brain barrier penetration and effective drug delivery in glioma therapy. Biomater Sci 2017; 5:1041-1050. [DOI: 10.1039/c7bm00133a] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A mustard-inspired delivery shuttle was constructed for enhanced blood–brain barrier penetration and effective drug delivery in glioma therapy.
Collapse
Affiliation(s)
- Nan Wang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Pei Sun
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Mingming Lv
- Department of Oral Maxillofacial-Head Neck Oncology
- Ninth People's Hospital
- Shanghai Jiao Tong University School of Medicine
- Shanghai Key Laboratory of Stomatology
- Shanghai 200011
| | - Gangsheng Tong
- Instrumental Analysis Center
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Xin Jin
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- P. R. China
| |
Collapse
|
6
|
Abe S, Yamamoto K, Kurata M, Abe-Suzuki S, Horii R, Akiyama F, Kitagawa M. Targeting MCM2 function as a novel strategy for the treatment of highly malignant breast tumors. Oncotarget 2016; 6:34892-909. [PMID: 26430873 PMCID: PMC4741497 DOI: 10.18632/oncotarget.5408] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 09/18/2015] [Indexed: 12/23/2022] Open
Abstract
Highly malignant tumors express high levels of the minichromosome maintenance 2 (MCM2) protein, which is associated with advanced tumor grade, advanced stage, and poor prognosis. In a previous study, we showed that Friend leukemia virus (FLV) envelope protein gp70 bound MCM2, impaired its nuclear translocation, and enhanced DNA-damage-induced apoptosis in FLV-infected hematopoietic cells when the cells expressed high levels of MCM2. Here, we show that MCM2 is highly expressed in clinical samples of invasive carcinoma of the breast, especially triple-negative breast cancer (TNBC), and in cancer stem cell (CSC) marker-positive breast cancer cells. To generate a cancer therapy model using gp70, we introduced the gp70 protein into the cytoplasm of murine breast cancer cells that express high levels of MCM2 by conjugating the protein transduction domain (PTD) of Hph-1 to gp70 (Hph- 1-gp70). Hph-1-gp70 was successfully transduced into the cytoplasm of breast cancer cells. The transduced protein enhanced the DNA damage-induced apoptosis of cancer cells in vitro and in vivo. Therefore, an MCM2-targeted strategy using Hph-1-gp70 treatment to induce DNA damage might be a successful therapy for highly malignant breast cancers such as TNBC and for the eradication of CSC-like cells from breast cancer tissue.
Collapse
Affiliation(s)
- Shinya Abe
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kouhei Yamamoto
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Morito Kurata
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shiho Abe-Suzuki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rie Horii
- Department of Pathology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Futoshi Akiyama
- Department of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masanobu Kitagawa
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
7
|
Yu J, Guo M, Wang T, Li X, Wang D, Wang X, Zhang Q, Wang L, Zhang Y, Zhao C, Feng B. Inhibition of cell proliferation, migration and invasion by a glioma-targeted fusion protein combining the p53 C terminus and MDM2-binding domain. Cell Prolif 2016; 49:79-89. [PMID: 26840447 DOI: 10.1111/cpr.12238] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 10/14/2015] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES The aim of this study was to develop multifunctional fusion proteins for targeting and delivering therapy elements into glioma cells. MATERIALS AND METHODS Multifunctional fusion proteins were expressed in Escherichia coli and purified using Ni-NTA resin affinity chromatography. Human glioma cells and primary astrocytes were used to analyse their functions. Targeting proteins location to glioma cells was observed by confocal microscopy. Effects of cell viability and proliferation were evaluated using the Cell Counting Kit 8 and colony formation assays. Glioma cell migration and invasion were assessed using transwell assays, and apoptosis was analysed by flow cytometry. In addition, changes in expression of proteins related to the cell cycle and apoptosis were determined by Western blotting. RESULTS The protein with highest bioactivity was GL1-riHA2-p53c+m-TAT (GHPc+mT), which combines glioma-targeting peptide GL1 (G), and C terminus (Pc) and mouse double minute domains (Pm) of p53, with the destabilizing lipid membrane peptide riHA2 (H) and cell-penetrating peptide TAT (T). The purified fusion protein was stable in cell culture medium and specifically targeted, and was internalized by, epidermal growth factor receptor (EGFR)-overexpressing glioma cells (U87ΔEGFR). It inhibited cell proliferation, migration and invasion, while flow cytometric analysis showed increased apoptosis. In addition, GHPc+mT caused significant changes in expression of proteins related to the cell cycle and apoptosis. CONCLUSION GHPc+mT is a multifunctional protein combining targeting, inhibition of glioma cell proliferation and induction of apoptosis, providing some potential to be developed into an effective protein drug delivery system for glioma therapy.
Collapse
Affiliation(s)
- Jiawen Yu
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China.,Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Meihua Guo
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Ting Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xiang Li
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Dan Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Xinying Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Qian Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Liang Wang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Yang Zhang
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| | - Chunhui Zhao
- College of Life Sciences, Liaoning Normal University, Dalian, 116029, China
| | - Bin Feng
- Department of Biotechnology, Dalian Medical University, Dalian, 116044, China
| |
Collapse
|
8
|
A recombinant trans-membrane protein hMnSOD–R9 inhibits the proliferation of cervical cancer cells in vitro. Mol Cell Biochem 2013; 385:79-86. [DOI: 10.1007/s11010-013-1816-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 09/13/2013] [Indexed: 12/30/2022]
|
9
|
KANNO HIROSHI, SATO HIDEMITSU, YOKOYAMA TAKAAKIRA, YOSHIZUMI TETSUYA, YAMADA SACHIKO. The VHL tumor suppressor protein regulates tumorigenicity of U87-derived glioma stem-like cells by inhibiting the JAK/STAT signaling pathway. Int J Oncol 2013; 42:881-6. [DOI: 10.3892/ijo.2013.1773] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/17/2012] [Indexed: 11/06/2022] Open
|