1
|
Cetin MH, Rieckmann T, Hoffer K, Riepen B, Christiansen S, Gatzemeier F, Feyerabend S, Schoof M, Schüller U, Petersen C, Mynarek M, Rothkamm K, Kriegs M, Struve N. G2 checkpoint targeting via Wee1 inhibition radiosensitizes EGFRvIII-positive glioblastoma cells. Radiat Oncol 2023; 18:19. [PMID: 36709315 PMCID: PMC9884419 DOI: 10.1186/s13014-023-02210-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The gene of the Epidermal growth factor receptor (EGFR) is one of the most frequently altered genes in glioblastoma (GBM), with deletions of exons 2-7 (EGFRvIII) being amongst the most common genomic mutations. EGFRvIII is heterogeneously expressed in GBM. We already showed that EGFRvIII expression has an impact on chemosensitivity, replication stress, and the DNA damage response. Wee1 kinase is a major regulator of the DNA damage induced G2 checkpoint. It is highly expressed in GBM and its overexpression is associated with poor prognosis. Since Wee1 inhibition can lead to radiosensitization of EGFRvIII-negative (EGFRvIII-) GBM cells, we asked, if Wee1 inhibition is sufficient to radiosensitize also EGFRvIII-positive (EGFRvIII+) GBM cells. METHODS We used the clinically relevant Wee1 inhibitor adavosertib and two pairs of isogenetic GBM cell lines with and without endogenous EGFRvIII expression exhibiting different TP53 status. Moreover, human GBM samples displaying heterogenous EGFRvIII expression were analyzed. Expression of Wee1 was assessed by Western blot and respectively immunohistochemistry. The impact of Wee1 inhibition in combination with irradiation on cell cycle and cell survival was analyzed by flow cytometry and colony formation assay. RESULTS Analysis of GBM cells and patient samples revealed a higher expression of Wee1 in EGFRvIII+ cells compared to their EGFRvIII- counterparts. Downregulation of EGFRvIII expression by siRNA resulted in a strong decrease in Wee1 expression. Wee1 inhibition efficiently abrogated radiation-induced G2-arrest and caused radiosensitization, without obvious differences between EGFRvIII- and EGFRvIII+ GBM cells. CONCLUSION We conclude that the inhibition of Wee1 is an effective targeting approach for the radiosensitization of both EGFRvIII- and EGFRvIII+ GBM cells and may therefore represent a promising new therapeutic option to increase response to radiotherapy.
Collapse
Affiliation(s)
- Meryem H. Cetin
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Thorsten Rieckmann
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany ,grid.13648.380000 0001 2180 3484 Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstantin Hoffer
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Britta Riepen
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Sabrina Christiansen
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Fruzsina Gatzemeier
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Simon Feyerabend
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Melanie Schoof
- grid.470174.1Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- grid.470174.1Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Cordula Petersen
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Martin Mynarek
- grid.13648.380000 0001 2180 3484Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany ,grid.13648.380000 0001 2180 3484Mildred-Scheel Cancer Career Center HaTriCs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Malte Kriegs
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Nina Struve
- grid.13648.380000 0001 2180 3484Department of Radiobiology & Radiation Oncology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany ,grid.13648.380000 0001 2180 3484Mildred-Scheel Cancer Career Center HaTriCs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
Struve N, Hoffer K, Weik AS, Riepen B, Krug L, Cetin MH, Burmester J, Ott L, Liebing J, Gatzemeier F, Müller-Goebel J, Gerbach M, Bußmann L, Parplys AC, Unger K, Mansour WY, Schüller U, Rieckmann T, Petersen C, Rothkamm K, Short SC, Kriegs M. Increased replication stress and R-loop accumulation in EGFRvIII-expressing glioblastoma present new therapeutic opportunities. Neurooncol Adv 2022; 4:vdab180. [PMID: 35274102 PMCID: PMC8903237 DOI: 10.1093/noajnl/vdab180] [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] [Indexed: 11/16/2022] Open
Abstract
Background The oncogene epidermal growth factor receptor variant III (EGFRvIII) is expressed in approximately one-third of all glioblastomas (GBMs). So far it is not clear if EGFRvIII expression induces replication stress in GBM cells, which might serve as a therapeutical target. Methods Isogenetic EGFRvIII− and EGFRvIII+ cell lines with endogenous EGFRvIII expression were used. Markers of oncogenic and replication stress such as γH2AX, RPA, 53BP1, ATR, and CHK1 were analyzed using western blot, immunofluorescence, and flow cytometry. The DNA fiber assay was performed to analyze replication, transcription was measured by incorporation of EU, and genomic instability was investigated by micronuclei and CGH-Array analysis. Immunohistochemistry staining was used to detect replication stress markers and R-loops in human GBM samples. Results EGFRvIII+ cells exhibit an activated replication stress response, increased spontaneous DNA damage, elevated levels of single-stranded DNA, and reduced DNA replication velocity, which are all indicative characteristics of replication stress. Furthermore, we show here that EGFRvIII expression is linked to increased genomic instability. EGFRvIII-expressing cells display elevated RNA synthesis and R-loop formation, which could also be confirmed in EGFRvIII-positive GBM patient samples. Targeting replication stress by irinotecan resulted in increased sensitivity of EGFRvIII+ cells. Conclusion This study demonstrates that EGFRvIII expression is associated with increased replication stress, R-loop accumulation, and genomic instability. This might contribute to intratumoral heterogeneity but may also be exploited for individualized therapy approaches.
Collapse
Affiliation(s)
- Nina Struve
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred-Scheel Cancer Career Center HATRICs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Konstantin Hoffer
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna-Sophie Weik
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Britta Riepen
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leonie Krug
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Meryem H Cetin
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasmin Burmester
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leonie Ott
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana Liebing
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Fruzsina Gatzemeier
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Justus Müller-Goebel
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mirja Gerbach
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lara Bußmann
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred-Scheel Cancer Career Center HATRICs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Radiobiology & Experimental Radiation, Germany
| | - Ann Christin Parplys
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiotherapy and Radiooncology, Philipps University, Marburg, Germany
| | - Kristian Unger
- Research Unit Radiation Cytogenetics, Helmholtz Zentrum München, Oberschleibheim, Germany
| | - Wael Y Mansour
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred-Scheel Cancer Career Center HATRICs4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Thorsten Rieckmann
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otolaryngology and Head and Neck Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Laboratory of Radiobiology & Experimental Radiation, Germany
| | - Cordula Petersen
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susan C Short
- Leeds Institute of Cancer and Pathology, St James's University Hospital, UK
| | - Malte Kriegs
- Department of Radiotherapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Xu Y, Shen M, Li Y, Sun Y, Teng Y, Wang Y, Duan Y. The synergic antitumor effects of paclitaxel and temozolomide co-loaded in mPEG-PLGA nanoparticles on glioblastoma cells. Oncotarget 2018; 7:20890-901. [PMID: 26956046 PMCID: PMC4991499 DOI: 10.18632/oncotarget.7896] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 02/20/2016] [Indexed: 12/13/2022] Open
Abstract
To get better chemotherapy efficacy, the optimal synergic effect of Paclitaxel (PTX) and Temozolomide (TMZ) on glioblastoma cells lines was investigated. A dual drug-loaded delivery system based on mPEG-PLGA nanoparticles (NPs) was developed to potentiate chemotherapy efficacy for glioblastoma. PTX/TMZ-NPs were prepared with double emulsification solvent evaporation method and exhibited a relatively uniform diameter of 206.3 ± 14.7 nm. The NPs showed sustained release character. Cytotoxicity assays showed the best synergistic effects were achieved when the weight ratios of PTX to TMZ were 1:5 and 1:100 on U87 and C6 cells, respectively. PTX/TMZ-NPs showed better inhibition effect to U87 and C6 cells than single drug NPs or free drugs mixture. PTX/TMZ-NPs (PTX: TMZ was 1:5(w/w)) significantly inhibited the tumor growth in the subcutaneous U87 mice model. These results indicate that coordinate administration of PTX and TMZ combined with NPs is an efficient method for glioblastoma.
Collapse
Affiliation(s)
- Yuanyuan Xu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, P. R. China
| | - Ming Shen
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, P. R. China
| | - Yiming Li
- Department of Ultrasound, Huashan Hospital, School of Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, P. R. China
| | - Yanwei Teng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, P. R. China
| | - Yi Wang
- Department of Ultrasound, Huashan Hospital, School of Medicine, Fudan University, Shanghai 200040, P. R. China
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, P. R. China
| |
Collapse
|
4
|
Winograd EK, Ciesielski MJ, Fenstermaker RA. Novel vaccines for glioblastoma: clinical update and perspective. Immunotherapy 2017; 8:1293-1308. [PMID: 27993092 DOI: 10.2217/imt-2016-0059] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Glioblastoma is the most common primary brain cancer. Aggressive treatment with surgery, radiation therapy and chemotherapy provides limited overall survival benefit. Glioblastomas have a formidable tumor microenvironment that is hostile to immunological effector cells and these cancers produce profound systemic immunosuppression. However, surgical resection of these tumors creates conditions that favor the use of immunotherapeutic strategies. Therefore, extensive surgical resection, when feasible, will remain part of the equation to provide an environment in which active specific immunotherapy has the greatest chance of working. Toward that end, a number of vaccination protocols are under investigation. Vaccines studied to date have produced cellular and humoral antitumor responses, but unequivocal clinical efficacy has yet to be demonstrated. In addition, focus is shifting toward the prospect of therapies involving vaccines in combination with immune checkpoint inhibitors and other immunomodulatory agents so that effector cells remain active against their targets systemically and within the tumor microenvironment.
Collapse
Affiliation(s)
- Evan K Winograd
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA
| | - Michael J Ciesielski
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA.,Department of Neurosurgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Robert A Fenstermaker
- Department of Neurosurgery, State University of New York at Buffalo, Jacobs School of Medicine & Biomedical Sciences, Buffalo, NY 14260, USA.,Department of Neurosurgery, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| |
Collapse
|
5
|
Ding L, Wang Q, Shen M, Sun Y, Zhang X, Huang C, Chen J, Li R, Duan Y. Thermoresponsive nanocomposite gel for local drug delivery to suppress the growth of glioma by inducing autophagy. Autophagy 2017; 13:1176-1190. [PMID: 28594260 DOI: 10.1080/15548627.2017.1320634] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Although the treatments of malignant glioma include surgery, radiotherapy and chemotherapy by oral drug administration, the prognosis of patients with glioma remains very poor. We developed a polyethylene glycol-dipalmitoylphosphatidyle- thanoiamine (mPEG-DPPE) calcium phosphate nanoparticles (NPs) injectable thermoresponsive hydrogel (nanocomposite gel) that could provide a sustained and local delivery of paclitaxel (PTX) and temozolomide (TMZ). In addition, the proportion of PTX and TMZ for the optimal synergistic antiglioma effect on C6 cells was determined to be 1:100 (w/w) by the Chou and Talalay method. Our results clearly indicated that the autophagy induced by PTX:TMZ NPs plays an important role in regulating tumor cell death, while autophagy inhibition dramatically reverses the antitumor effect of PTX:TMZ NPs, suggesting that antiproliferative autophagy occurs in response to PTX:TMZ NPs treatment. The antitumor efficacy of the PTX:TMZ NP-loaded gel was evaluated in situ using C6 tumor-bearing rats, and the PTX:TMZ NP-loaded gel exhibited superior antitumor performance. The antitumor effects of the nanocomposite gel in vivo were shown to correlate with autophagic cell death in this study. The in vivo results further confirmed the advantages of such a strategy. The present study may provide evidence supporting the development of nanomedicine for potential clinical application.
Collapse
Affiliation(s)
- Li Ding
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Qi Wang
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Ming Shen
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Ying Sun
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Xiangyu Zhang
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Can Huang
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Jianhua Chen
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Rongxin Li
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| | - Yourong Duan
- a State Key Laboratory of Oncogenes and Related Genes , School of Biomedical Engineering, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University , Shanghai , China
| |
Collapse
|
6
|
The effects of CD147 on the cell proliferation, apoptosis, invasion, and angiogenesis in glioma. Neurol Sci 2016; 38:129-136. [PMID: 27761842 DOI: 10.1007/s10072-016-2727-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
Abstract
To analyze the effects of extracellular matrix metalloproteinase inducer (CD147) on glioma proliferation, apoptosis, invasion, and angiogenesis. Tissue samples were obtained from 101 glioma cases while normal brain tissues were obtained from 30 brain injury cases. Immunohistochemical assay was performed to detect the expressions of CD147, CD34, and VEGF in tissue samples. QRT-PCR was performed to detect the relative expression of CD147 mRNA in human glioma cell lines. CD147 siRNA was transfected into glioma cell line U251. Cell proliferation, apoptosis, invasion, and angiogenesis were tested by MTT, flow cytometry, Transwell assay, and vasculogenic mimicry assay, respectively. Expressions of relative proteins were analyzed with western blot. CD147 was positively expressed with the percentage of 0, 37.5, 44.8, 67.9, and 85.7 % in normal tissues and glioma tissues with WHO grades I-IV, respectively, and the scores of MVDand VEGF were associated with the expression of CD147. CD147 was significantly upregulated in the human glioma cell lines (P < 0.05). Downregulated the expression of CD147 suppressed cell proliferation, blocked cell cycle, induced apoptosis, inhibited cell invasion and angiogenesis in glioma cells in vitro. The expression of CD147 was significantly associated with WHO tumor grade and angiogenesis; silencing of CD147 contributed to inhibition of glioma proliferation, invasion, and angiogenesis. Our study provided firm evidence that CD 147 is a potential glioma target for anti-angiogenic therapies.
Collapse
|
7
|
Albrecht V, Richter A, Pfeiffer S, Gebauer M, Lindner S, Gieser E, Schüller U, Schichor C, Gildehaus FJ, Bartenstein P, Tonn JC, Skerra A, Glass R. Anticalins directed against the fibronectin extra domain B as diagnostic tracers for glioblastomas. Int J Cancer 2015; 138:1269-80. [PMID: 26421425 DOI: 10.1002/ijc.29874] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 08/31/2015] [Indexed: 12/22/2022]
Abstract
The standard of care for diagnosis and therapy monitoring of gliomas is magnetic resonance imaging (MRI), which however, provides only an indirect and incomplete representation of the tumor mass, offers limited information for patient stratification according to WHO-grades and may insufficiently indicate tumor relapse after antiangiogenic therapy. Anticalins are alternative binding proteins obtained via combinatorial protein design from the human lipocalin scaffold that offer novel diagnostic reagents for histology and imaging applications. Here, the Anticalins N7A, N7E and N9B, which possess exquisite specificity and affinity for oncofetal fibronectin carrying the extra domain B (ED-B), a well-known proangiogenic extracellular matrix protein, were applied for immunohistochemical studies. When investigating ED-B expression in biopsies from 41 patients with confirmed gliomas of WHO grades I to IV, or in non-neoplastic brain samples, we found that Anticalins specifically detect ED-B in primary glioblastoma multiforme (GBM; WHO IV) but not in tumors of lower histopathological grade or in tumor-free brain. In primary GBM samples, ED-B specific Anticalins locate to fibronectin-rich perivascular areas that are associated with angiogenesis. Anticalins specifically detect ED-B both in fixed tumor specimen and on vital cells, as evidenced by cytofluorometry. Beyond that, we labeled an Anticalin with the γ-emitter (123) I and demonstrated specific binding to GBM-tissue samples using in vitro autoradiography. Overall, our data indicate that ED-B specific Anticalins are useful tools for the diagnosis of primary GBM and related angiogenic sites, presenting them as promising tracers for molecular tumor imaging.
Collapse
Affiliation(s)
- Valerie Albrecht
- Department of Neurosurgery, Neurosurgical Research, University of Munich, Munich, Germany
| | - Antonia Richter
- Munich Center for Integrated Protein Science (CIPS-M) & Lehrstuhl für Biologische Chemie, Technische Universität München, Freising-Weihenstephan, Germany
| | - Sarah Pfeiffer
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Michaela Gebauer
- Munich Center for Integrated Protein Science (CIPS-M) & Lehrstuhl für Biologische Chemie, Technische Universität München, Freising-Weihenstephan, Germany
| | - Simon Lindner
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Eugenie Gieser
- Department of Neurosurgery, Neurosurgical Research, University of Munich, Munich, Germany
| | - Ulrich Schüller
- Center for Neuropathology and Prion Research, University of Munich, Munich, Germany
| | - Christian Schichor
- Department of Neurosurgery, Neurosurgical Research, University of Munich, Munich, Germany
| | | | - Peter Bartenstein
- Department of Nuclear Medicine, University of Munich, Munich, Germany
| | - Jörg-Christian Tonn
- Department of Neurosurgery, Neurosurgical Research, University of Munich, Munich, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) & Lehrstuhl für Biologische Chemie, Technische Universität München, Freising-Weihenstephan, Germany
| | - Rainer Glass
- Department of Neurosurgery, Neurosurgical Research, University of Munich, Munich, Germany
| |
Collapse
|
8
|
Wang X, Zhang K, Chen X, Zhao C, Sun Z. Epilysin is overexpressed in glioblastoma and related to clinical outcome of patients. Med Oncol 2014; 32:363. [PMID: 25429835 DOI: 10.1007/s12032-014-0363-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
As the newest identified member of the matrix metalloproteinase (MMP) family, the expression pattern and function of epilysin (MMP-28) are still not well understood. Although epilysin was found to play an evolutionarily conserved role in neural development, the expression and function of epilysin in malignant glioma are unknown. Therefore, the aim of the present study was to quantitatively evaluate the expression level of epilysin in glioblastoma (GBM) and its association with clinical outcome of patients. For this purpose, a total of 216 GBM specimens and 31 normal brain specimens were collected in the present study. Expression level of epilysin was determined by immunohistochemistry assay and immunoreactivity score system. MGMT promoter methylation and IDH1/2 mutation status in GBM were also evaluated. Results showed that the positive rate of epilysin staining in GBM was significantly elevated compared with that in normal brain. Positive epilysin staining was associated with low KPS score, unmethylated MGMT promoter and wild-type IDH. Kaplan-Meier analysis showed that patients with GBM of positive epilysin staining were more likely to have unfavorable overall survival. Multivariate analysis revealed that epilysin was an independent and significant prognostic marker of GBM. These results proved for the first time that epilysin expression was significantly elevated in GBM and can be potentially used to predict prognosis in patients with GBM.
Collapse
Affiliation(s)
- Xuepeng Wang
- China-Japan Union Hospital, Jilin University, Changchun, People's Republic of China
| | | | | | | | | |
Collapse
|
9
|
Vecchio D, Daga A, Carra E, Marubbi D, Baio G, Neumaier CE, Vagge S, Corvò R, Pia Brisigotti M, Louis Ravetti J, Zunino A, Poggi A, Mascelli S, Raso A, Frosina G. Predictability, efficacy and safety of radiosensitization of glioblastoma-initiating cells by the ATM inhibitor KU-60019. Int J Cancer 2014; 135:479-491. [PMID: 24443327 DOI: 10.1002/ijc.28680] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 11/21/2013] [Accepted: 11/25/2013] [Indexed: 12/12/2022]
Abstract
We have previously shown that pharmacological inhibition of ataxia telangiectasia mutated (ATM) protein sensitizes glioblastoma-initiating cells (GICs) to ionizing radiation (IR). Herein, we report the experimental conditions to overcome GIC radioresistance in vitro using the specific ATM inhibitor KU-60019, two major determinants of the tumor response to this drug and the absence of toxicity of this treatment in vitro and in vivo. Repeated treatments with KU-60019 followed by IR substantially delayed GIC proliferation in vitro and even eradicated radioresistant cells, whereas GIC treated with vehicle plus radiation recovered early and expanded. The tumor response to the drug occurred under a cutoff level of expression of TP53 and over a cutoff level of expression of phosphatidylinositol 3-kinase (PI3K). No increased clastogenicity or point mutagenicity was induced by KU-60019 plus radiation when compared to vehicle plus radiation. No significant histological changes to the brain or other organs were observed after prolonged infusion into the brain of KU-60019 at millimolar concentrations. Taken together, these findings suggest that GIC-driven tumors with low expression of TP53 and high expression of PI3K might be effectively and safely radiosensitized by KU-60019.
Collapse
Affiliation(s)
- Donatella Vecchio
- Mutagenesis Unit, IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Thirant C, Gavard J, Junier MP, Chneiweiss H. Critical multiple angiogenic factors secreted by glioblastoma stem-like cells underline the need for combinatorial anti-angiogenic therapeutic strategies. Proteomics Clin Appl 2014; 7:79-90. [PMID: 23229792 DOI: 10.1002/prca.201200102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/31/2012] [Accepted: 11/14/2012] [Indexed: 01/06/2023]
Abstract
Glioblastomas are the most frequent adult primary brain tumors that still remain fatal despite major clinical efforts. As in other solid tumors, populations of glioblastoma stem-like cells (GSCs) endowed with tumor initiating and therapeutic resistance properties have been identified. Glioblastomas are highly vascularized tumors resulting in a rich dialog between GSCs and endothelial cells. In one direction, endothelial cells and their secreted proteins are able to sustain GSC properties while, in turn, GSCs can promote neoangiogenesis, modulate endothelial cell functions and may even transdifferentiate into endothelial cells. Accordingly, targeting tumor vasculature seems a promising issue despite incomplete and transient results obtained from anti-vascular endothelial growth factor therapeutic trials. Recent findings of novel GSC-secreted molecules with pro-angiogenic properties (Semaphorin 3A, hepatoma-derived growth factor) open the path to the design of a concerted attack of glioblastoma vasculature that could overcome the development of resistance to single-targeted therapies while keeping away the toxicity of the treatments.
Collapse
Affiliation(s)
- Cécile Thirant
- Leukemia and Stem Cell Biology Laboratory, Department of Hematological Medicine, Rayne Institute, King's College London, London, UK
| | | | | | | |
Collapse
|
11
|
Ni S, Fan X, Wang J, Qi H, Li X. Biodegradable Implants Efficiently Deliver Combination of Paclitaxel and Temozolomide to Glioma C6 Cancer Cells In Vitro. Ann Biomed Eng 2013; 42:214-21. [DOI: 10.1007/s10439-013-0903-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 08/27/2013] [Indexed: 11/29/2022]
|
12
|
Yang M, Yuan Y, Zhang H, Yan M, Wang S, Feng F, Ji P, Li Y, Li B, Gao G, Zhao J, Wang L. Prognostic significance of CD147 in patients with glioblastoma. J Neurooncol 2013; 115:19-26. [PMID: 23925827 DOI: 10.1007/s11060-013-1207-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 07/15/2013] [Indexed: 12/21/2022]
Abstract
CD147, also known as extracellular matrix metalloproteinase inducer, is a widely distributed cell surface glycoprotein that belongs to the immunoglobulin superfamily. CD147 has been proved to be enriched on the surface of many tumor cells, promoting tumor growth, invasion and metastasis by its stimulation effect on adjacent fibroblasts to produce matrix metalloproteinases. In this study, we aimed to explore the expression pattern of CD147 in glioblastoma (GBM) and investigate whether it could be used to assess subsequent prognosis of patients. For that, we recruited a total of 206 patients with pathologically confirmed GBM and 36 normal control brain tissue specimens. The expression of CD147 in GBM and normal tissues was investigated by immunohistochemistry assay. Genetic factors including MGMT and IDH1 mutation were also investigated to justify the prognostic significance of CD147. Results showed that CD147 expression was increased in GBM compared with that in normal tissues. Kaplan-Meier analysis showed that increased CD147 expression was associated with poor overall survival of patients with GBM. Moreover, Cox's proportional hazards model revealed that CD147 expression was an independent and significant prognostic marker of overall survival in GBM patients. These results proved that CD147 expression was relatively abundant in GBM and can be potentially used to predict prognosis and treatment response in GBM patients.
Collapse
Affiliation(s)
- Min Yang
- Institute of Orthopaedics and Traumatology of PLA of China, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Resistance to two heterologous neurotropic oncolytic viruses, Semliki Forest virus and vaccinia virus, in experimental glioma. J Virol 2012; 87:2363-6. [PMID: 23221568 DOI: 10.1128/jvi.01609-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Attenuated Semliki Forest virus (SFV) may be suitable for targeting malignant glioma due to its natural neurotropism, but its replication in brain tumor cells may be restricted by innate antiviral defenses. We attempted to facilitate SFV replication in glioma cells by combining it with vaccinia virus, which is capable of antagonizing such defenses. Surprisingly, we found parenchymal mouse brain tumors to be refractory to both viruses. Also, vaccinia virus appears to be sensitive to SFV-induced antiviral interference.
Collapse
|
14
|
Lowenstein PR, Castro MG. Pushing the limits of glioma resection using electrophysiologic brain mapping. J Clin Oncol 2012; 30:2437-40. [PMID: 22529267 DOI: 10.1200/jco.2011.40.6959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
15
|
Shirazi HA, Grimm S, Raizer J, Mehta MP. Combined modality approaches in the management of adult glioblastoma. Front Oncol 2011; 1:36. [PMID: 22655242 PMCID: PMC3356104 DOI: 10.3389/fonc.2011.00036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 09/29/2011] [Indexed: 11/16/2022] Open
Abstract
Over the past two decades, management of newly diagnosed glioblastoma has undergone significant evolution. While surgery has long been a mainstay of management for this disease, and while radiotherapy has a proven survival role, initial efforts at radiotherapy dose escalation, use of radiosurgery, brachytherapy, and altered fractionation did not improve patient survival. Recently, multiple modality therapy integrating maximal safe resection, postoperative radiation, and new systemic therapies have resulted in improved patient outcomes compared with older regimens utilizing surgery and postoperative radiation alone. Numerous trials are currently underway investigating the combination of surgery, radiation, and systemic therapy with targeted agents to find ways to further improve outcomes for adults with glioblastoma.
Collapse
Affiliation(s)
- Haider A. Shirazi
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Sean Grimm
- Department of Neurology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Jeffrey Raizer
- Department of Neurology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| | - Minesh P. Mehta
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern UniversityChicago, IL, USA
| |
Collapse
|