1
|
Dehghan N, Mousavikia SN, Qasempour Y, Azimian H. Radiation-induced senescence in glioblastoma: An overview of the mechanisms and eradication strategies. Life Sci 2024; 359:123218. [PMID: 39510171 DOI: 10.1016/j.lfs.2024.123218] [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: 08/22/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
Radiotherapy as a treatment method for glioblastoma is limited due to the intrinsic apoptosis resistance mechanisms of the tumor. Administration of higher radiation doses contributes to toxicities in normal tissues and organs at risk, like optic chiasma. Cellular senescence represents an alternative mechanism to apoptosis following radiotherapy in glioblastoma, occurring in both normal and neoplastic cells. Although it impedes the growth of tumors and sustains cells in their cycle, it can also act as a cause of tumor development and recurrence following treatment. In this review, we discuss detailed insights into the significance of radiation-induced senescence in glioblastoma and the underlying mechanisms that lead to radioresistance. We also discuss senescence biomarkers and the role of senescence-associated secretory phenotype (SASP) in tumor recurrence. Finally, we review the studies that have administered potential interventions to eradicate or inhibit senescent cells in glioblastoma after treatment with radiation.
Collapse
Affiliation(s)
- Neda Dehghan
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Nasibeh Mousavikia
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Younes Qasempour
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Franco MS, Raulefs S, Schilling D, Combs SE, Schmid TE. Impact of Radiation on Invasion and Migration of Glioma In Vitro and In Vivo. Cancers (Basel) 2024; 16:3900. [PMID: 39682088 DOI: 10.3390/cancers16233900] [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: 10/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Glioblastoma (GBM) constitutes the most common primary brain tumor and it remains incurable despite therapeutic advances. The high infiltration/invasion potential of GBM cells is considered to be one of the reasons for the inevitable recurrence of the disease. Radiotherapy (RT) is part of the standard care for patients with GBM, and its benefits on overall survival are extensively reported. However, numerous preclinical studies show that X-ray irradiation can enhance the motility of GBM cells. In the present review, we bring together state-of-the-art research on the impact of radiation on GBM cell motility. The mechanisms through which irradiation impacts the brain tumor microenvironment and the tumor cells themselves, leading to more aggressive/invasive tumors, are described. Finally, we summarize potential pharmacological strategies to overcome this problem. Clinical data validating the occurrence of these processes are urgently needed as they could be of great value for patient outcomes. With this comprehensive review, we expect to highlight the need for methods which allow for monitoring the post-irradiation invasive behavior of GBM in patients.
Collapse
Affiliation(s)
- Marina Santiago Franco
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Susanne Raulefs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Daniela Schilling
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| | - Stephanie E Combs
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Thomas E Schmid
- School of Medicine and Health, Department of Radiation Oncology, TUM University Hospital, Technical University of Munich, 81675 Munich, Germany
- Institute of Radiation Medicine, Helmholtz Munich, 85764 Neuherberg, Germany
| |
Collapse
|
3
|
Frascogna C, Mottareale R, La Verde G, Arrichiello C, Muto P, Netti PA, Pugliese M, Panzetta V. Role of the mechanical microenvironment on CD-44 expression of breast adenocarcinoma in response to radiotherapy. Sci Rep 2024; 14:391. [PMID: 38172135 PMCID: PMC10764959 DOI: 10.1038/s41598-023-50473-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/20/2023] [Indexed: 01/05/2024] Open
Abstract
The biological effects of ionizing radiation are exploited in the clinical practice of radiotherapy to destroy tumour cells while sparing the surrounding normal tissue. While most of the radiotherapy research focused on DNA damage and repair, recently a great attention is going to cells' interactions with the mechanical microenvironment of both malignant and healthy tissues after exposure. In fact, the stiffness of the extracellular matrix can modify cells' motility and spreading through the modulation of transmembrane proteins and surface receptors' expression, such as CD-44. CD-44 receptor has held much interest also in targeted-therapy due to its affinity with hyaluronic acid, which can be used to functionalize biodegradable nanoparticles loaded with chemotherapy drugs for targeted therapy. We evaluated changes in CD-44 expression in two mammary carcinoma cell lines (MCF10A and MDA-MB-231) after exposure to X-ray (2 or 10 Gy). To explore the role of the mechanical microenvironment, we mimicked tissues' stiffness with polyacrylamide's substrates producing two different elastic modulus values (0.5 and 15 kPa). We measured a dose dependent increase in CD-44 relative expression in tumour cells cultured in a stiffer microenvironment. These findings highlight a crucial connection between the mechanical properties of the cell's surroundings and the post-radiotherapy expression of surface receptors.
Collapse
Affiliation(s)
- Crescenzo Frascogna
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| | - Rocco Mottareale
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
| | - Giuseppe La Verde
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy
- Istituto Nazionale di Fisica Nucleare, INFN Sezione di Napoli, Via Cinthia Ed. 6, 80126, Naples, Italy
| | - Cecilia Arrichiello
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 53, 80131, Naples, Italy
| | - Paolo Muto
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione "G. Pascale", Via Semmola, 53, 80131, Naples, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials CRIB, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| | - Mariagabriella Pugliese
- Department of Physics "E. Pancini", University of Naples Federico II, Via Cinthia, 80126, Naples, Italy.
- Istituto Nazionale di Fisica Nucleare, INFN Sezione di Napoli, Via Cinthia Ed. 6, 80126, Naples, Italy.
| | - Valeria Panzetta
- Center for Advanced Biomaterials for Healthcare @CRIB, Italian Institute of Technology, Largo Barsanti e Matteucci 53, 80125, Naples, Italy
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
- Interdisciplinary Research Centre On Biomaterials CRIB, University of Naples Federico II, Piazzale Vincenzo Tecchio, 80125, Naples, Italy
| |
Collapse
|
4
|
Li Z, Ge H, Xie Y, Zhang Y, Zhao X, Sun W, Song M. Luteolin inhibits angiogenesis and enhances radiotherapy sensitivity of laryngeal cancer via downregulating Integrin β1. Tissue Cell 2023; 85:102235. [PMID: 37826960 DOI: 10.1016/j.tice.2023.102235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/05/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023]
Abstract
AIM To demonstrate the role and mechanism of luteolin in radio-sensitization and angiogenesis of laryngeal cancer. METHODS Firstly, we analyzed the cytotoxicity of Luteolin and radiation sensitive cytotoxicity through CCK8, and selected subsequent radiation doses and Luteolin concentrations. Next, we further analyzed the effects of Luteolin on radiation sensitivity and neovascularization of laryngeal cancer, and conducted CCK8, plate cloning, and angiogenesis experiments, respectively. At the same time, the effects of individual treatment and combination treatment on the expression of Integrin β1 and VEGFA were analyzed through immunofluorescence analysis. We also analyzed the regulation of Integrin β1 protein expression by Luteolin through Western blot. To investigate the mechanism of Integrin β1, we transfected overexpressed and silenced Integrin β1 vectors and analyzed the role of Integrin β1 in Luteolin enhancing radiation sensitivity of laryngeal cancer by repeating the above experiments. We have also constructed an in vivo subcutaneous tumor transplantation model to further validate the cell experimental results. The expression of Integrin, KI67, VEGFA, and CD31 was analyzed through Western blot and immunohistochemistry experiments. RESULTS Radiation inhibited cell proliferation and decreased Integrin β1 expression, and increased the radiosensitivity through inhibiting cell proliferation, and inhibit angiogenesis during radiation. Overexpression of Integrin β1 weakened radiotherapy sensitivity on the basis of cells treated with combined administration. Integrin β1 is considered as the downstream molecule of luteolin, participating in radiosensitivity of luteolin to FaDu cells. Animal experiments also demonstrated that luteolin strengthened tumor suppression and anti-angiogenesis during radiation via Integrin β1. CONCLUSION In summary, our results manifested that radio-sensitivity effect of luteolin depended on downregulating Integrin β1 in laryngocarcinoma.
Collapse
Affiliation(s)
- Zhen Li
- Department of Otorhinolaryngology, Yantaishan Hospital, Yantai, Shandong, China
| | - Hongzhou Ge
- Department of Otorhinolaryngology, Qingdao Traditional Chinese Medicine Hospital (Qingdao Hiser Hospital) Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, Shandong, China
| | - Yonggang Xie
- Department of Anesthesiology, The Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yueqin Zhang
- Department of Otorhinolaryngology, Yantaishan Hospital, Yantai, Shandong, China
| | - Xiaoyan Zhao
- Department of Otorhinolaryngology, Yantaishan Hospital, Yantai, Shandong, China
| | - Wen Sun
- Department of Otorhinolaryngology, Yantaishan Hospital, Yantai, Shandong, China
| | - Meiyan Song
- Administrative Department, Yantaishan Hospital, Yantai, Shandong, China.
| |
Collapse
|
5
|
Elser M, Vehlow A, Juratli TA, Cordes N. Simultaneous inhibition of discoidin domain receptor 1 and integrin αVβ3 radiosensitizes human glioblastoma cells. Am J Cancer Res 2023; 13:4597-4612. [PMID: 37970361 PMCID: PMC10636682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 08/13/2023] [Indexed: 11/17/2023] Open
Abstract
Glioblastomas (GBM) are the most common primary brain tumors in adults and associated with poor clinical outcomes due to therapy resistances and destructive growth. Interactions of cancer cells with the extracellular matrix (ECM) play a pivotal role in therapy resistances and tumor progression. In this study, we investigate the functional dependencies between the discoidin domain receptor 1 (DDR1) and the integrin family of cell adhesion molecules for the radioresponse of human glioblastoma cells. By means of an RNA interference screen on DDR1 and all known integrin subunits, we identified co-targeting of DDR1/integrin β3 to most efficiently reduce clonogenicity, enhance cellular radiosensitivity and diminish repair of DNA double strand breaks (DSB). Simultaneous pharmacological inhibition of DDR1 with DDR1-IN-1 and of integrins αVβ3/αVβ5 with cilengitide resulted in confirmatory data in a panel of 2D grown glioblastoma cultures and 3D gliospheres. Mechanistically, we found that key DNA repair proteins ATM and DNA-PK are altered upon DDR1/integrin αVβ3/integrin αVβ5 inhibition, suggesting a link to DNA repair mechanisms. In sum, the radioresistance of human glioblastoma cells can effectively be declined by co-deactivation of DDR1, integrin αVβ3 and integrin αVβ5.
Collapse
Affiliation(s)
- Marc Elser
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden01307 Dresden, Germany
| | - Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden01307 Dresden, Germany
| | - Tareq A Juratli
- Department of Neurosurgery, Division of Neuro-Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden01307 Dresden, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden, German Cancer Research Center69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden01307 Dresden, Germany
| |
Collapse
|
6
|
Whitehead CA, Fang H, Su H, Morokoff AP, Kaye AH, Hanssen E, Nowell CJ, Drummond KJ, Greening DW, Vella LJ, Mantamadiotis T, Stylli SS. Small extracellular vesicles promote invadopodia activity in glioblastoma cells in a therapy-dependent manner. Cell Oncol (Dordr) 2023; 46:909-931. [PMID: 37014551 PMCID: PMC10356899 DOI: 10.1007/s13402-023-00786-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2023] [Indexed: 04/05/2023] Open
Abstract
PURPOSE The therapeutic efficacy of radiotherapy/temozolomide treatment for glioblastoma (GBM) is limited by the augmented invasiveness mediated by invadopodia activity of surviving GBM cells. As yet, however the underlying mechanisms remain poorly understood. Due to their ability to transport oncogenic material between cells, small extracellular vesicles (sEVs) have emerged as key mediators of tumour progression. We hypothesize that the sustained growth and invasion of cancer cells depends on bidirectional sEV-mediated cell-cell communication. METHODS Invadopodia assays and zymography gels were used to examine the invadopodia activity capacity of GBM cells. Differential ultracentrifugation was utilized to isolate sEVs from conditioned medium and proteomic analyses were conducted on both GBM cell lines and their sEVs to determine the cargo present within the sEVs. In addition, the impact of radiotherapy and temozolomide treatment of GBM cells was studied. RESULTS We found that GBM cells form active invadopodia and secrete sEVs containing the matrix metalloproteinase MMP-2. Subsequent proteomic studies revealed the presence of an invadopodia-related protein sEV cargo and that sEVs from highly invadopodia active GBM cells (LN229) increase invadopodia activity in sEV recipient GBM cells. We also found that GBM cells displayed increases in invadopodia activity and sEV secretion post radiation/temozolomide treatment. Together, these data reveal a relationship between invadopodia and sEV composition/secretion/uptake in promoting the invasiveness of GBM cells. CONCLUSIONS Our data indicate that sEVs secreted by GBM cells can facilitate tumour invasion by promoting invadopodia activity in recipient cells, which may be enhanced by treatment with radio-chemotherapy. The transfer of pro-invasive cargos may yield important insights into the functional capacity of sEVs in invadopodia.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
| | - Haoyun Fang
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Huaqi Su
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Neurosurgery, Hadassah Hebrew University Medical Centre, Jerusalem, Israel
| | - Eric Hanssen
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3010, Australia
- Advanced Microscopy Facility, The Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Cameron J Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, 3052, Australia
| | - Katharine J Drummond
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Melbourne, VIC, Australia
- Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia
| | - Laura J Vella
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Centre for Stem Cell Systems, The University of Melbourne, Parkville, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Stanley S Stylli
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia.
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Level 5, Clinical Sciences Building, Parkville, VIC, 3050, Australia.
| |
Collapse
|
7
|
Jones D, Whitehead CA, Dinevska M, Widodo SS, Furst LM, Morokoff AP, Kaye AH, Drummond KJ, Mantamadiotis T, Stylli SS. Repurposing FDA-approved drugs as inhibitors of therapy-induced invadopodia activity in glioblastoma cells. Mol Cell Biochem 2023; 478:1251-1267. [PMID: 36302993 PMCID: PMC10164021 DOI: 10.1007/s11010-022-04584-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 10/11/2022] [Indexed: 11/28/2022]
Abstract
Glioblastoma (GBM) is the most prevalent primary central nervous system tumour in adults. The lethality of GBM lies in its highly invasive, infiltrative, and neurologically destructive nature resulting in treatment failure, tumour recurrence and death. Even with current standard of care treatment with surgery, radiotherapy and chemotherapy, surviving tumour cells invade throughout the brain. We have previously shown that this invasive phenotype is facilitated by actin-rich, membrane-based structures known as invadopodia. The formation and matrix degrading activity of invadopodia is enhanced in GBM cells that survive treatment. Drug repurposing provides a means of identifying new therapeutic applications for existing drugs without the need for discovery or development and the associated time for clinical implementation. We investigate several FDA-approved agents for their ability to act as both cytotoxic agents in reducing cell viability and as 'anti-invadopodia' agents in GBM cell lines. Based on their cytotoxicity profile, three agents were selected, bortezomib, everolimus and fludarabine, to test their effect on GBM cell invasion. All three drugs reduced radiation/temozolomide-induced invadopodia activity, in addition to reducing GBM cell viability. These drugs demonstrate efficacious properties warranting further investigation with the potential to be implemented as part of the treatment regime for GBM.
Collapse
Affiliation(s)
- Dylan Jones
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Clarissa A Whitehead
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Marija Dinevska
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Samuel S Widodo
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Liam M Furst
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew P Morokoff
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Andrew H Kaye
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Hadassah University Medical Centre, 91120, Jerusalem, Israel
| | - Katharine J Drummond
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Theo Mantamadiotis
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
- Department of Microbiology and Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Stanley S Stylli
- Level 5, Clinical Sciences Building, Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
- Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia.
| |
Collapse
|
8
|
Guerron A, Phan HT, Peñaloza-Arias C, Brambilla D, Roullin VG, Giasson S. Selectively triggered cell detachment from poly(N-isopropylacrylamide) microgel functionalized substrates. Colloids Surf B Biointerfaces 2022. [DOI: 10.1016/j.colsurfb.2022.112699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
9
|
Oishi T, Koizumi S, Kurozumi K. Molecular Mechanisms and Clinical Challenges of Glioma Invasion. Brain Sci 2022; 12:brainsci12020291. [PMID: 35204054 PMCID: PMC8870089 DOI: 10.3390/brainsci12020291] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/17/2022] Open
Abstract
Glioma is the most common primary brain tumor, and its prognosis is poor. Glioma cells are highly invasive to the brain parenchyma. It is difficult to achieve complete resection due to the nature of the brain tissue, and tumors that invade the parenchyma often recur. The invasiveness of tumor cells has been studied from various aspects, and the related molecular mechanisms are gradually becoming clear. Cell adhesion factors and extracellular matrix factors have a strong influence on glioma invasion. The molecular mechanisms that enhance the invasiveness of glioma stem cells, which have been investigated in recent years, have also been clarified. In addition, it has been discussed from both basic and clinical perspectives that current therapies can alter the invasiveness of tumors, and there is a need to develop therapeutic approaches to glioma invasion in the future. In this review, we will summarize the factors that influence the invasiveness of glioma based on the environment of tumor cells and tissues, and describe the impact of the treatment of glioma on invasion in terms of molecular biology, and the novel therapies for invasion that are currently being developed.
Collapse
|
10
|
Liang S, Zhou G, Hu W. Research Progress of Heavy Ion Radiotherapy for Non-Small-Cell Lung Cancer. Int J Mol Sci 2022; 23:2316. [PMID: 35216430 PMCID: PMC8876478 DOI: 10.3390/ijms23042316] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 02/05/2023] Open
Abstract
Non-small-cell lung cancer (NSCLC) has a high incidence and poses a serious threat to human health. However, the treatment outcomes of concurrent chemoradiotherapy for non-small-cell lung cancer are still unsatisfactory, especially for high grade lesions. As a new cancer treatment, heavy ion radiotherapy has shown promising efficacy and safety in the treatment of non-small-cell lung cancer. This article discusses the clinical progress of heavy ion radiotherapy in the treatment of non-small-cell lung cancer mainly from the different cancer stages, the different doses of heavy ion beams, and the patient's individual factors, and explores the deficiency of heavy ion radiotherapy in the treatment of non-small-cell lung cancer and the directions of future research, in order to provide reference for the wider and better application of heavy ion radiotherapy in the future.
Collapse
Affiliation(s)
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China;
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China;
| |
Collapse
|
11
|
Vehlow A, Cordes N. Growth factor receptor and β1 integrin signaling differentially regulate basal clonogenicity and radiation survival of fibroblasts via a modulation of cell cycling. In Vitro Cell Dev Biol Anim 2022; 58:169-178. [PMID: 35194763 PMCID: PMC8901520 DOI: 10.1007/s11626-022-00656-z] [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: 06/30/2021] [Accepted: 02/02/2022] [Indexed: 11/30/2022]
Abstract
Cell adhesion to extracellular matrix proteins mediates resistance to radio- and chemotherapy by activating integrin signaling. In addition, mutual and cooperative interactions between integrin and growth factor receptor signaling contribute to the cellular radiation response. Here, we investigate to which extend the crosstalk between β1 integrins and growth factor receptor signaling determines the cellular radiation response of fibroblasts by assessing clonogenic survival and cell cycling. By utilizing growth factor signaling competent and either β1 integrin wildtype GD25β1A fibroblasts or β1 integrin mutant, signaling incompetent GD25β1B fibroblasts, we show basal clonogenic survival to depend on growth factor receptor but not integrin signaling. Our data further suggest the cooperation between β1 integrins and growth factor receptors to be critical for enhancing the radiation-induced G2/M cell cycle block leading to improved clonogenic radiation survival. By pharmacological inhibition of EGFR and PI3K, we additionally show that the essential contribution of EGFR signaling to radiogenic G2/M cell cycle arrest depends on the co-activation of the β1 integrin signaling axis, but occurs independent of PI3K. Taken together, elucidation of the signaling circuitry underlying the EGFR/β1 integrin crosstalk may support the development of advanced molecular targeted therapies for radiation oncology.
Collapse
Affiliation(s)
- Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307, Dresden, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 41, 01307, Dresden, Germany. .,Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstr. 74, PF 50, 01307, Dresden, Germany. .,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Bautzner Landstr. 400, 01328, Dresden, Germany. .,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany, Im Neuenheimer Feld 280, 69192, Heidelberg, Germany.
| |
Collapse
|
12
|
Heffernan JM, McNamara JB, Vernon BL, Mehta S, Sirianni RW. PNJ scaffolds promote microenvironmental regulation of glioblastoma stem-like cell enrichment and radioresistance. Biomater Sci 2022; 10:819-833. [PMID: 34994746 PMCID: PMC8939461 DOI: 10.1039/d0bm01169j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glioblastoma (GBM) brain tumors contain a subpopulation of self-renewing multipotent Glioblastoma stem-like cells (GSCs) that are believed to drive the near inevitable recurrence of GBM. We previously engineered temperature responsive scaffolds based on the polymer poly(N-isopropylacrylamide-co-Jeffamine M-1000 acrylamide) (PNJ) for the purpose of enriching GSCs in vitro from patient-derived samples. Here, we used PNJ scaffolds to study microenvironmental regulation of self-renewal and radiation response in patient-derived GSCs representing classical and proneural subtypes. GSC self-renewal was regulated by the composition of PNJ scaffolds and varied with cell type. PNJ scaffolds protected against radiation-induced cell death, particularly in conditions that also promoted GSC self-renewal. Additionally, cells cultured in PNJ scaffolds exhibited increased expression of the transcription factor HIF2α, which was not observed in neurosphere culture, providing a potential mechanistic basis for differences in radio-resistance. Differences in PNJ regulation of HIF2α in irradiated and untreated conditions also offered evidence of stem plasticity. These data show PNJ scaffolds provide a unique biomaterial for evaluating dynamic microenvironmental regulation of GSC self-renewal, radioresistance, and stem plasticity.
Collapse
Affiliation(s)
- John M. Heffernan
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA, Sonoran Biosciences, 1048 E Knight Ln, Tempe, AZ, USA
| | - James B. McNamara
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, Arizona 85721, USA
| | - Brent L. Vernon
- School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA
| | - Shwetal Mehta
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA
| | - Rachael W. Sirianni
- Ivy Brain Tumor Center, Barrow Neurological Institute, 350 W Thomas Rd, Phoenix, AZ 85013, USA, School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ 85287, USA, Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| |
Collapse
|
13
|
Djuzenova CS, Fischer T, Katzer A, Sisario D, Korsa T, Steussloff G, Sukhorukov VL, Flentje M. Opposite effects of the triple target (DNA-PK/PI3K/mTOR) inhibitor PI-103 on the radiation sensitivity of glioblastoma cell lines proficient and deficient in DNA-PKcs. BMC Cancer 2021; 21:1201. [PMID: 34763650 PMCID: PMC8582108 DOI: 10.1186/s12885-021-08930-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 10/28/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Radiotherapy is routinely used to combat glioblastoma (GBM). However, the treatment efficacy is often limited by the radioresistance of GBM cells. METHODS Two GBM lines MO59K and MO59J, differing in intrinsic radiosensitivity and mutational status of DNA-PK and ATM, were analyzed regarding their response to DNA-PK/PI3K/mTOR inhibition by PI-103 in combination with radiation. To this end we assessed colony-forming ability, induction and repair of DNA damage by γH2AX and 53BP1, expression of marker proteins, including those belonging to NHEJ and HR repair pathways, degree of apoptosis, autophagy, and cell cycle alterations. RESULTS We found that PI-103 radiosensitized MO59K cells but, surprisingly, it induced radiation resistance in MO59J cells. Treatment of MO59K cells with PI-103 lead to protraction of the DNA damage repair as compared to drug-free irradiated cells. In PI-103-treated and irradiated MO59J cells the foci numbers of both proteins was higher than in the drug-free samples, but a large portion of DNA damage was quickly repaired. Another cell line-specific difference includes diminished expression of p53 in MO59J cells, which was further reduced by PI-103. Additionally, PI-103-treated MO59K cells exhibited an increased expression of the apoptosis marker cleaved PARP and increased subG1 fraction. Moreover, irradiation induced a strong G2 arrest in MO59J cells (~ 80% vs. ~ 50% in MO59K), which was, however, partially reduced in the presence of PI-103. In contrast, treatment with PI-103 increased the G2 fraction in irradiated MO59K cells. CONCLUSIONS The triple-target inhibitor PI-103 exerted radiosensitization on MO59K cells, but, unexpectedly, caused radioresistance in the MO59J line, lacking DNA-PK. The difference is most likely due to low expression of the DNA-PK substrate p53 in MO59J cells, which was further reduced by PI-103. This led to less apoptosis as compared to drug-free MO59J cells and enhanced survival via partially abolished cell-cycle arrest. The findings suggest that the lack of DNA-PK-dependent NHEJ in MO59J line might be compensated by DNA-PK independent DSB repair via a yet unknown mechanism.
Collapse
Affiliation(s)
- Cholpon S Djuzenova
- Department of Radiation Oncology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany.
| | - Thomas Fischer
- Department of Radiation Oncology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| | - Astrid Katzer
- Department of Radiation Oncology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| | - Dmitri Sisario
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Tessa Korsa
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Gudrun Steussloff
- Department of Radiation Oncology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| | - Vladimir L Sukhorukov
- Department of Biotechnology and Biophysics, University of Würzburg, Würzburg, Germany
| | - Michael Flentje
- Department of Radiation Oncology, University Hospital of Würzburg, Josef-Schneider-Strasse 11, 97080, Würzburg, Germany
| |
Collapse
|
14
|
Cytoskeleton Response to Ionizing Radiation: A Brief Review on Adhesion and Migration Effects. Biomedicines 2021; 9:biomedicines9091102. [PMID: 34572287 PMCID: PMC8465203 DOI: 10.3390/biomedicines9091102] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 12/27/2022] Open
Abstract
The cytoskeleton is involved in several biological processes, including adhesion, motility, and intracellular transport. Alterations in the cytoskeletal components (actin filaments, intermediate filaments, and microtubules) are strictly correlated to several diseases, such as cancer. Furthermore, alterations in the cytoskeletal structure can lead to anomalies in cells’ properties and increase their invasiveness. This review aims to analyse several studies which have examined the alteration of the cell cytoskeleton induced by ionizing radiations. In particular, the radiation effects on the actin cytoskeleton, cell adhesion, and migration have been considered to gain a deeper knowledge of the biophysical properties of the cell. In fact, the results found in the analysed works can not only aid in developing new diagnostic tools but also improve the current cancer treatments.
Collapse
|
15
|
Serafim RB, da Silva P, Cardoso C, Di Cristofaro LFM, Netto RP, de Almeida R, Navegante G, Storti CB, de Sousa JF, de Souza FC, Panepucci R, Moreira CG, Penna LS, Silva WA, Valente V. Expression Profiling of Glioblastoma Cell Lines Reveals Novel Extracellular Matrix-Receptor Genes Correlated With the Responsiveness of Glioma Patients to Ionizing Radiation. Front Oncol 2021; 11:668090. [PMID: 34211843 PMCID: PMC8240593 DOI: 10.3389/fonc.2021.668090] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 04/30/2021] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most lethal and frequent type of brain tumor, leading patients to death in approximately 14 months after diagnosis. GBM treatment consists in surgical removal followed by radio and chemotherapy. However, tumors commonly relapse and the treatment promotes only a slight increase in patient survival. Thus, uncovering the cellular mechanisms involved in GBM resistance is of utmost interest, and the use of cell lines has been shown to be an extremely important tool. In this work, the exploration of RNAseq data from different GBM cell lines revealed different expression signatures, distinctly correlated with the behavior of GBM cell lines regarding proliferation indexes and radio-resistance. U87MG and U138MG cells, which presented expressively reduced proliferation and increased radio-resistance, showed a particular expression signature encompassing enrichment in many extracellular matrix (ECM) and receptor genes. Contrasting, U251MG and T98G cells, that presented higher proliferation and sensibility to radiation, exhibited distinct signatures revealing consistent enrichments for DNA repair processes and although several genes from the ECM-receptor pathway showed up-regulation, enrichments for this pathway were not detected. The ECM-receptor is a master regulatory pathway that is known to impact several cellular processes including: survival, proliferation, migration, invasion, and DNA damage signaling and repair, corroborating the associations we found. Furthermore, searches to The Cancer Genome Atlas (TCGA) repository revealed prognostic correlations with glioma patients for the majority of genes highlighted in the signatures and led to the identification of 31 ECM-receptor genes individually correlated with radiation responsiveness. Interestingly, we observed an association between the number of upregulated genes and survivability greater than 5 years after diagnosis, where almost all the patients that presented 21 or more upregulated genes were deceased before 5 years. Altogether our findings suggest the clinical relevance of ECM-receptor genes signature found here for radiotherapy decision and as biomarkers of glioma prognosis.
Collapse
Affiliation(s)
- Rodolfo Bortolozo Serafim
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Patrick da Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Cibele Cardoso
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | | | - Renato Petitto Netto
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Rodrigo de Almeida
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Geovana Navegante
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Camila Baldin Storti
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Juliana Ferreira de Sousa
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Felipe Canto de Souza
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Rodrigo Panepucci
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | | | - Larissa Siqueira Penna
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Wilson Araujo Silva
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Valeria Valente
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
- Center for Cell-Based Therapy (CTC), Regional Blood Center of Ribeirão Preto, Ribeirão Preto, Brazil
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, Brazil
| |
Collapse
|
16
|
Huang J, Zhang L, Wan D, Zhou L, Zheng S, Lin S, Qiao Y. Extracellular matrix and its therapeutic potential for cancer treatment. Signal Transduct Target Ther 2021; 6:153. [PMID: 33888679 PMCID: PMC8062524 DOI: 10.1038/s41392-021-00544-0] [Citation(s) in RCA: 442] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 02/17/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is one of the major components of tumors that plays multiple crucial roles, including mechanical support, modulation of the microenvironment, and a source of signaling molecules. The quantity and cross-linking status of ECM components are major factors determining tissue stiffness. During tumorigenesis, the interplay between cancer cells and the tumor microenvironment (TME) often results in the stiffness of the ECM, leading to aberrant mechanotransduction and further malignant transformation. Therefore, a comprehensive understanding of ECM dysregulation in the TME would contribute to the discovery of promising therapeutic targets for cancer treatment. Herein, we summarized the knowledge concerning the following: (1) major ECM constituents and their functions in both normal and malignant conditions; (2) the interplay between cancer cells and the ECM in the TME; (3) key receptors for mechanotransduction and their alteration during carcinogenesis; and (4) the current therapeutic strategies targeting aberrant ECM for cancer treatment.
Collapse
Affiliation(s)
- Jiacheng Huang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Lele Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Dalong Wan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lin Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China
| | - Shengzhang Lin
- School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310000, China.
| | - Yiting Qiao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, 310003, China.
- Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou, 310003, China.
- Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, 310003, China.
| |
Collapse
|
17
|
Berg TJ, Marques C, Pantazopoulou V, Johansson E, von Stedingk K, Lindgren D, Jeannot P, Pietras EJ, Bergström T, Swartling FJ, Governa V, Bengzon J, Belting M, Axelson H, Squatrito M, Pietras A. The Irradiated Brain Microenvironment Supports Glioma Stemness and Survival via Astrocyte-Derived Transglutaminase 2. Cancer Res 2021; 81:2101-2115. [PMID: 33483373 DOI: 10.1158/0008-5472.can-20-1785] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/02/2020] [Accepted: 01/19/2021] [Indexed: 11/16/2022]
Abstract
The tumor microenvironment plays an essential role in supporting glioma stemness and radioresistance. Following radiotherapy, recurrent gliomas form in an irradiated microenvironment. Here we report that astrocytes, when pre-irradiated, increase stemness and survival of cocultured glioma cells. Tumor-naïve brains increased reactive astrocytes in response to radiation, and mice subjected to radiation prior to implantation of glioma cells developed more aggressive tumors. Extracellular matrix derived from irradiated astrocytes were found to be a major driver of this phenotype and astrocyte-derived transglutaminase 2 (TGM2) was identified as a promoter of glioma stemness and radioresistance. TGM2 levels increased after radiation in vivo and in recurrent human glioma, and TGM2 inhibitors abrogated glioma stemness and survival. These data suggest that irradiation of the brain results in the formation of a tumor-supportive microenvironment. Therapeutic targeting of radiation-induced, astrocyte-derived extracellular matrix proteins may enhance the efficacy of standard-of-care radiotherapy by reducing stemness in glioma. SIGNIFICANCE: These findings presented here indicate that radiotherapy can result in a tumor-supportive microenvironment, the targeting of which may be necessary to overcome tumor cell therapeutic resistance and recurrence. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2101/F1.large.jpg.
Collapse
Affiliation(s)
- Tracy J Berg
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Carolina Marques
- Seve Ballesteros Foundation Brain Tumor group, CNIO, Madrid, Spain
| | - Vasiliki Pantazopoulou
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Elinn Johansson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kristoffer von Stedingk
- Department of Pediatrics, Clinical Sciences Lund, Lund University, Lund, Sweden.,Department of Oncogenomics, M1-131 Academic Medical Center University of Amsterdam, Amsterdam, the Netherlands
| | - David Lindgren
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Pauline Jeannot
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Elin J Pietras
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Tobias Bergström
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Fredrik J Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Valeria Governa
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Johan Bengzon
- Division of Neurosurgery, Department of Clinical Sciences, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mattias Belting
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden.,Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| | - Håkan Axelson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Alexander Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
18
|
New Avenues in Radiotherapy of Glioblastoma: from Bench to Bedside. Curr Treat Options Neurol 2020. [DOI: 10.1007/s11940-020-00654-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Kohata T, Ito S, Masuda T, Furuta T, Nakada M, Ohtsuki S. Laminin Subunit Alpha-4 and Osteopontin Are Glioblastoma-Selective Secreted Proteins That Are Increased in the Cerebrospinal Fluid of Glioblastoma Patients. J Proteome Res 2020; 19:3542-3553. [PMID: 32628487 DOI: 10.1021/acs.jproteome.0c00415] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor in adults. The purpose of the present study was to identify GBM cell-selective secreted proteins by analyzing conditioned media (CM) from GBM, breast, and colon cancer cell lines using sequential window acquisition of all theoretical spectra-mass spectrometry (SWATH-MS) and targeted proteomics. We identified 2371 proteins in the CM from GBM and the other cancer cell lines. Among the proteins identified, 15 showed significantly higher expression in the CM from GBM cell lines than in those from other cancer cell lines. These GBM-selective secreted proteins were further quantified in the cerebrospinal fluid (CSF) from patients with GBM. Laminin subunit alpha-4 (LAMA4) and osteopontin (OPN) had increased expression levels in the CSF from GBM patients compared to those from non-brain tumor patients. In addition, the areas under the curves in a receiver operating characteristic analysis of LAMA4 and OPN were greater than 0.9, allowing for discrimination of GBM patients from non-brain tumor patients. The CSF levels of LAMA4 and OPN were also significantly correlated with the GBM tumor volume. These results suggest that LAMA4 and OPN are secreted from GBM cells into the CSF and appear to be candidates as diagnostic markers and therapeutic targets for GBM.
Collapse
Affiliation(s)
- Tomohiro Kohata
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shingo Ito
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Tokyo, Japan
| | - Takeshi Masuda
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Tokyo, Japan
| | - Takuya Furuta
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan.,Department of Neurosurgery, Kanazawa University, Kanazawa, Japan
| | | | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,AMED-CREST, Tokyo, Japan
| |
Collapse
|
20
|
Panzetta V, La Verde G, Pugliese M, Artiola V, Arrichiello C, Muto P, La Commara M, Netti PA, Fusco S. Adhesion and Migration Response to Radiation Therapy of Mammary Epithelial and Adenocarcinoma Cells Interacting with Different Stiffness Substrates. Cancers (Basel) 2020; 12:E1170. [PMID: 32384675 PMCID: PMC7281676 DOI: 10.3390/cancers12051170] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/27/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022] Open
Abstract
The structural and mechanical properties of the microenvironmental context have a profound impact on cancer cell motility, tumor invasion, and metastasis formation. In fact, cells react to their mechanical environment modulating their adhesion, cytoskeleton organization, changes of shape, and, consequently, the dynamics of their motility. In order to elucidate the role of extracellular matrix stiffness as a driving force in cancer cell motility/invasion and the effects of ionizing radiations on these processes, we evaluated adhesion and migration as biophysical properties of two different mammary cell lines, over a range of pathophysiological stiffness (1-13 kPa) in a control condition and after the exposure to two different X-ray doses (2 and 10 Gy, photon beams). We concluded that the microenvironment mimicking the normal mechanics of healthy tissue has a radioprotective role on both cell lines, preventing cell motility and invasion. Supraphysiological extracellular matrix stiffness promoted tumor cell motility instead, but also had a normalizing effect on the response to radiation of tumor cells, lowering their migratory capability. This work lays the foundation for exploiting the extracellular matrix-mediated mechanism underlying the response of healthy and tumor cells to radiation treatments and opens new frontiers in the diagnostic and therapeutic use of radiotherapy.
Collapse
Affiliation(s)
- Valeria Panzetta
- Centro di Ricerca Interdipartimentale sui Biomateriali, Università degli Studi di Napoli Federico II, Piazzale Tecchio 80, 80125 Napoli, Italy;
- Centre for Advanced Biomaterial for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Napoli, Italy
| | - Giuseppe La Verde
- Istituto Nazionale di Fisica Nucleare, INFN sezione di Napoli, Via Cinthia ed. 6, 80126 Napoli, Italy; (G.L.V.); (M.P.); (M.L.C.)
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Montesano 49, 80131 Napoli, Italy
| | - Mariagabriella Pugliese
- Istituto Nazionale di Fisica Nucleare, INFN sezione di Napoli, Via Cinthia ed. 6, 80126 Napoli, Italy; (G.L.V.); (M.P.); (M.L.C.)
- Dipartimento di Fisica “Ettore Pancini”, Università degli Studi di Napoli Federico II, Via Cinthia ed. 6, 80126 Napoli, Italy;
| | - Valeria Artiola
- Dipartimento di Fisica “Ettore Pancini”, Università degli Studi di Napoli Federico II, Via Cinthia ed. 6, 80126 Napoli, Italy;
| | - Cecilia Arrichiello
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, Via Semmola, 53, 80131 Naples, Italy; (C.A.); (P.M.)
| | - Paolo Muto
- Radiotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, Via Semmola, 53, 80131 Naples, Italy; (C.A.); (P.M.)
| | - Marco La Commara
- Istituto Nazionale di Fisica Nucleare, INFN sezione di Napoli, Via Cinthia ed. 6, 80126 Napoli, Italy; (G.L.V.); (M.P.); (M.L.C.)
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via Montesano 49, 80131 Napoli, Italy
| | - Paolo A. Netti
- Centro di Ricerca Interdipartimentale sui Biomateriali, Università degli Studi di Napoli Federico II, Piazzale Tecchio 80, 80125 Napoli, Italy;
- Centre for Advanced Biomaterial for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Napoli, Italy
| | - Sabato Fusco
- Centro di Ricerca Interdipartimentale sui Biomateriali, Università degli Studi di Napoli Federico II, Piazzale Tecchio 80, 80125 Napoli, Italy;
- Centre for Advanced Biomaterial for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Napoli, Italy
| |
Collapse
|
21
|
Zhu C, Kong Z, Wang B, Cheng W, Wu A, Meng X. ITGB3/CD61: a hub modulator and target in the tumor microenvironment. Am J Transl Res 2019; 11:7195-7208. [PMID: 31934272 PMCID: PMC6943458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 11/24/2019] [Indexed: 06/10/2023]
Abstract
β3 integrin (ITGB3), also known as CD61 or GP3A, is one of the most widely studied components in the integrin family. As an adhesion receptor on the cell surface, ITGB3 participates in reprogramming tumor metabolism, shaping the stromal and immune microenvironment, facilitating epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (End-MT) and maintaining tumor stemness, etc. Recent studies proposed various intervention strategies against ITGB3 and have achieved promising outcomes in several types of tumor. Here, we review the adaption response and cellular crosstalk in the tumor microenvironment mediated by ITGB3, as well as its upstream and downstream signaling pathways. Lastly, we focus on the inhibitors of ITGB3, ultimately indicating that ITGB3 is a promising target in the tumor microenvironment.
Collapse
Affiliation(s)
- Chen Zhu
- Department of Neurosurgery, The First Hospital of China Medical UniversityShenyang, Liaoning, China
| | - Ziqing Kong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical UniversityShenyang, Liaoning, China
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical UniversityShenyang, Liaoning, China
| | - Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical UniversityShenyang, Liaoning, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical UniversityShenyang, Liaoning, China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical UniversityShenyang, Liaoning, China
| |
Collapse
|
22
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
23
|
Eke I, Makinde AY, Aryankalayil MJ, Reedy JL, Citrin DE, Chopra S, Ahmed MM, Coleman CN. Long-term Tumor Adaptation after Radiotherapy: Therapeutic Implications for Targeting Integrins in Prostate Cancer. Mol Cancer Res 2018; 16:1855-1864. [PMID: 30042176 PMCID: PMC6279542 DOI: 10.1158/1541-7786.mcr-18-0232] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/24/2018] [Accepted: 07/06/2018] [Indexed: 11/16/2022]
Abstract
Adaptation of tumor cells to radiotherapy induces changes that are actionable by molecular targeted agents and immunotherapy. This report demonstrates that radiation-induced changes in integrin expression can be targeted 2 months later. Integrins are transmembrane cell adhesion molecules that are essential for cancer cell survival and proliferation. To analyze the short- and long-term effects of radiation on the integrin expression, prostate cancer cells (DU145, PC3, and LNCaP) were cultured in a 3D extracellular matrix and irradiated with either a single dose of radiation (2-10 Gy) or a multifractionated regimen (2-10 fractions of 1 Gy). Whole human genome microarrays, immunoblotting, immunoprecipitation assays, and immunofluorescence staining of integrins were performed. The results were confirmed in a prostate cancer xenograft model system. Interestingly, β1 and β4 integrins (ITGB1 and ITGB4) were upregulated after radiation in vitro and in vivo. This overexpression lasted for more than 2 months and was dose dependent. Moreover, radiation-induced upregulation of β1 and β4 integrin resulted in significantly increased tumor cell death after treatment with inhibitory antibodies. Combined, these findings indicate that long-term tumor adaptation to radiation can result in an increased susceptibility of surviving cancer cells to molecular targeted therapy due to a radiation-induced overexpression of the target. IMPLICATIONS: Radiation induces dose- and schedule-dependent adaptive changes that are targetable for an extended time; thus suggesting radiotherapy as a unique strategy to orchestrate molecular processes, thereby providing new radiation-drug treatment options within precision cancer medicine.
Collapse
Affiliation(s)
- Iris Eke
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| | - Adeola Y Makinde
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jessica L Reedy
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sunita Chopra
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mansoor M Ahmed
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Radiation Research Program, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
24
|
Whitehead CA, Nguyen HPT, Morokoff AP, Luwor RB, Paradiso L, Kaye AH, Mantamadiotis T, Stylli SS. Inhibition of Radiation and Temozolomide-Induced Invadopodia Activity in Glioma Cells Using FDA-Approved Drugs. Transl Oncol 2018; 11:1406-1418. [PMID: 30219696 PMCID: PMC6140414 DOI: 10.1016/j.tranon.2018.08.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/27/2018] [Accepted: 08/27/2018] [Indexed: 12/31/2022] Open
Abstract
The most common primary central nervous system tumor in adults is the glioblastoma multiforme (GBM). The highly invasive nature of GBM cells is a significant factor resulting in the inevitable tumor recurrence and poor patient prognosis. Tumor cells utilize structures known as invadopodia to faciliate their invasive phenotype. In this study, utilizing an array of techniques, including gelatin matrix degradation assays, we show that GBM cell lines can form functional gelatin matrix degrading invadopodia and secrete matrix metalloproteinase 2 (MMP-2), a known invadopodia-associated matrix-degrading enzyme. Furthermore, these cellular activities were augmented in cells that survived radiotherapy and temozolomide treatment, indicating that surviving cells may possess a more invasive phenotype posttherapy. We performed a screen of FDA-approved agents not previously used for treating GBM patients with the aim of investigating their "anti-invadopodia" and cytotoxic effects in GBM cell lines and identified a number that reduced cell viability, as well as agents which also reduced invadopodia activity. Importantly, two of these, pacilitaxel and vinorelbine tartrate, reduced radiation/temozolomide-induced invadopodia activity. Our data demonstrate the value of testing previously approved drugs (repurposing) as potential adjuvant agents for the treatment of GBM patients to reduce invadopodia activity, inhibit GBM cell invasion, and potentially improve patient outcome.
Collapse
Affiliation(s)
- Clarissa A Whitehead
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Hong P T Nguyen
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andrew P Morokoff
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Rodney B Luwor
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Lucia Paradiso
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andrew H Kaye
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Theo Mantamadiotis
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Microbiology & Immunology, School of Biomedical Sciences, The University of Melbourne, Parkville VIC 3010, Victoria, Australia
| | - Stanley S Stylli
- Department of Surgery, The University of Melbourne, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia; Department of Neurosurgery, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia.
| |
Collapse
|
25
|
Wank M, Schilling D, Schmid TE, Meyer B, Gempt J, Barz M, Schlegel J, Liesche F, Kessel KA, Wiestler B, Bette S, Zimmer C, Combs SE. Human Glioma Migration and Infiltration Properties as a Target for Personalized Radiation Medicine. Cancers (Basel) 2018; 10:cancers10110456. [PMID: 30463322 PMCID: PMC6266328 DOI: 10.3390/cancers10110456] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 01/28/2023] Open
Abstract
Gliomas are primary brain tumors that present the majority of malignant adult brain tumors. Gliomas are subdivided into low- and high-grade tumors. Despite extensive research in recent years, the prognosis of malignant glioma patients remains poor. This is caused by naturally highly infiltrative capacities as well as high levels of radio- and chemoresistance. Additionally, it was shown that low linear energy transfer (LET) irradiation enhances migration and invasion of several glioma entities which might counteract today’s treatment concepts. However, this finding is discussed controversially. In the era of personalized medicine, this controversial data might be attributed to the patient-specific heterogeneity that ultimately could be used for treatment. Thus, current developments in glioma therapy should be seen in the context of intrinsic and radiation-enhanced migration and invasion. Due to the natural heterogeneity of glioma cells and different radiation responses, a personalized radiation treatment concept is suggested and alternative radiation concepts are discussed.
Collapse
Affiliation(s)
- Michaela Wank
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| | - Daniela Schilling
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Thomas E Schmid
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Bernhard Meyer
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Jens Gempt
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Melanie Barz
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
| | - Jürgen Schlegel
- Department of Neuropathology, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Friederike Liesche
- Department of Neuropathology, Technical University of Munich (TUM), 81675 Munich, Germany.
| | - Kerstin A Kessel
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| | - Benedikt Wiestler
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Stefanie Bette
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Claus Zimmer
- Department of Neuroradiology, Klinikum rechts der Isar, Technische Universität München, 81675 Munich, Germany.
| | - Stephanie E Combs
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, 85764 Neuherberg, Germany.
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, 81675 Munich, Germany.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site Munich, 81675 Munich, Germany.
| |
Collapse
|
26
|
Swenson S, Minea RO, Tuan CD, Thein TZ, Chen TC, Markland FS. A Novel Venom-Derived Peptide for Brachytherapy of Glioblastoma: Preclinical Studies in Mice. Molecules 2018; 23:molecules23112918. [PMID: 30413113 PMCID: PMC6278533 DOI: 10.3390/molecules23112918] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/01/2018] [Accepted: 11/03/2018] [Indexed: 11/16/2022] Open
Abstract
We developed a bacterial expression system to produce a recombinant disintegrin, vicrostatin (VCN), whose structure is based on a natural disintegrin isolated from southern copperhead snake venom. Our goal is to develop VCN for potential clinical translation as an anti-cancer agent. VCN is a peptide of 69 amino acids with a single tyrosine residue. We have employed VCN as integrin-targeted radionuclide therapy (brachytherapy) for treatment of glioblastoma (GBM, glioma). GBM is a deadly brain cancer that doesn't discriminate between sexes and knows no age limit. We established that the tyrosine residue in VCN can be radioiodinated with full retention of bioactivity. 131I-VCN was utilized for integrin-targeted radionuclide therapy using mouse models of glioma. The combination of radioiodinated VCN plus temozolomide (a DNA alkylating agent) significantly prolonged survival of glioma-bearing mice. We also obtained similar results using an immunocompetent mouse model and a murine glioma cell line. In summary, as demonstrated in studies reported here we have shown that VCN as targeted radionuclide therapy for GBM has significant translational potential for therapy of this deadly disease.
Collapse
Affiliation(s)
- Steve Swenson
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90089, USA.
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA 90089, USA.
| | - Radu O Minea
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA 90089, USA.
| | - Cao Duc Tuan
- Faculty of Pharmacy, Haiphong University of Medicine and Pharmacy, Haiphong, Vietnam.
| | - Thu-Zan Thein
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA 90089, USA.
| | - Thomas C Chen
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA 90089, USA.
| | - Francis S Markland
- Department of Biochemistry and Molecular Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
27
|
Hasslacher S, Schneele L, Stroh S, Langhans J, Zeiler K, Kattner P, Karpel-Massler G, Siegelin MD, Schneider M, Zhou S, Grunert M, Halatsch ME, Nonnenmacher L, Debatin KM, Westhoff MA. Inhibition of PI3K signalling increases the efficiency of radiotherapy in glioblastoma cells. Int J Oncol 2018; 53:1881-1896. [PMID: 30132519 PMCID: PMC6192725 DOI: 10.3892/ijo.2018.4528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma, the most common primary brain tumour, is also considered one of the most lethal cancers per se. It is highly refractory to therapeutic intervention, as highlighted by the mean patient survival of only 15 months, despite an aggressive treatment approach, consisting of maximal safe surgical resection, followed by radio- and chemotherapy. Radiotherapy, in particular, can have effects on the surviving fractions of tumour cells, which are considered adverse to the desired clinical outcome: It can induce increased cellular proliferation, as well as enhanced invasion. In this study, we established that differentiated glioblastoma cells alter their DNA repair response following repeated exposure to radiation and, therefore, high single-dose irradiation (SD-IR) is not a good surrogate marker for fractionated dose irradiation (FD-IR), as used in clinical practice. Integrating irradiation into a combination therapy approach, we then investigated whether the pharmacological inhibition of PI3K signalling, the most abundantly activated survival cascade in glioblastoma, enhances the efficacy of radiotherapy. Of note, treatment with GDC-0941, which blocks PI3K-mediated signalling, did not enhance cell death upon irradiation, but both treatment modalities functioned synergistically to reduce the total cell number. Furthermore, GDC-0941 not only prevented the radiation-induced increase in the motility of the differentiated cells, but further reduced their speed below that of untreated cells. Therefore, combining radiotherapy with the pharmacological inhibition of PI3K signalling is a potentially promising approach for the treatment of glioblastoma, as it can reduce the unwanted effects on the surviving fraction of tumour cells.
Collapse
Affiliation(s)
- Sebastian Hasslacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Lukas Schneele
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Sebastien Stroh
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Julia Langhans
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Katharina Zeiler
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Patricia Kattner
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Matthias Schneider
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Shaoxia Zhou
- Department of Clinical Chemistry, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Michael Grunert
- Department of Radiology, German Armed Forces Hospital of Ulm, D-89081 Ulm, Germany
| | - Marc-Eric Halatsch
- Department of Neurosurgery, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Lisa Nonnenmacher
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, D-89075 Ulm, Germany
| |
Collapse
|
28
|
Wank M, Schilling D, Reindl J, Meyer B, Gempt J, Motov S, Alexander F, Wilkens JJ, Schlegel J, Schmid TE, Combs SE. Evaluation of radiation-related invasion in primary patient-derived glioma cells and validation with established cell lines: impact of different radiation qualities with differing LET. J Neurooncol 2018; 139:583-590. [PMID: 29882045 DOI: 10.1007/s11060-018-2923-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/05/2018] [Indexed: 11/29/2022]
Abstract
PURPOSE Glioblastoma multiforme (GBM) is the most common primary brain tumor and has a very poor overall prognosis. Multimodal treatment is still inefficient and one main reason is the invasive nature of GBM cells, enabling the tumor cells to escape from the treatment area causing tumor progression. This experimental study describes the effect of low- and high-LET irradiation on the invasion of primary GBM cells with a validation in established cell systems. METHODS Seven patient derived primary GBM as well as three established cell lines (LN229, LN18 and U87) were used in this study. Invasion was investigated using Matrigel® coated transwell chambers. Irradiation was performed with low- (X-ray) and high-LET (alpha particles) radiation. The colony formation assay was chosen to determine the corresponding alpha particle dose equivalent to the X-ray dose. RESULTS 4 Gy X-ray irradiation increased the invasive potential of six patient derived GBM cells as well as two of the established lines. In contrast, alpha particle irradiation with an equivalent dose of 1.3 Gy did not show any effect on the invasive behavior. The findings were validated with established cell lines. CONCLUSION Our results show that in contrast to low-LET irradiation high-LET irradiation does not enhance the invasion of established and primary glioblastoma cell lines. We therefore suggest that high-LET irradiation could become an alternative treatment option. To fully exploit the benefits of high-LET irradiation concerning the invasion of GBM further molecular studies should be performed.
Collapse
Affiliation(s)
- M Wank
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Oberschleißheim, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site, Munich, Germany
| | - D Schilling
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Oberschleißheim, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site, Munich, Germany
| | - J Reindl
- Institute for Applied Physics and Metrology, Bundeswehr University Munich, Neubiberg, Germany
| | - B Meyer
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - J Gempt
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - S Motov
- Department of Neurosurgery, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - F Alexander
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - J J Wilkens
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - J Schlegel
- Department of Neuropathology, Technical University of Munich (TUM), Munich, Germany
| | - T E Schmid
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Oberschleißheim, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - S E Combs
- Institute of Innovative Radiotherapy (iRT), Department of Radiation Sciences (DRS), Helmholtz Zentrum München, Oberschleißheim, Germany. .,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany. .,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partner Site, Munich, Germany.
| |
Collapse
|
29
|
Abstract
Radiotherapy remains one of the corner stones in the treatment of various malignancies and often leads to an improvement in overall survival. Nonetheless, pre-clinical evidence indicates that radiation can entail pro-metastatic effects via multiple pathways. Via direct actions on cancer cells and indirect actions on the tumor microenvironment, radiation has the potential to enhance epithelial-to-mesenchymal transition, invasion, migration, angiogenesis and metastasis. However, the data remains ambiguous and clinical observations that unequivocally prove these findings are lacking. In this review we discuss the pre-clinical and clinical data on the local and systemic effect of irradiation on the metastatic process with an emphasis on the molecular pathways involved.
Collapse
|
30
|
Adhesion- and stress-related adaptation of glioma radiochemoresistance is circumvented by β1 integrin/JNK co-targeting. Oncotarget 2018; 8:49224-49237. [PMID: 28514757 PMCID: PMC5564763 DOI: 10.18632/oncotarget.17480] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/12/2017] [Indexed: 11/25/2022] Open
Abstract
Resistance of cancer stem-like and cancer tumor bulk cells to radiochemotherapy and destructive infiltration of the brain fundamentally influence the treatment efficiency to cure of patients suffering from Glioblastoma (GBM). The interplay of adhesion and stress-related signaling and activation of bypass cascades that counteract therapeutic approaches remain to be identified in GBM cells. We here show that combined inhibition of the adhesion receptor β1 integrin and the stress-mediator c-Jun N-terminal kinase (JNK) induces radiosensitization and blocks invasion in stem-like and patient-derived GBM cultures as well as in GBM cell lines. In vivo, this treatment approach not only significantly delays tumor growth but also increases median survival of orthotopic, radiochemotherapy-treated GBM mice. Both, in vitro and in vivo, effects seen with β1 integrin/JNK co-inhibition are superior to the monotherapy. Mechanistically, the in vitro radiosensitization provoked by β1 integrin/JNK targeting is caused by defective DNA repair associated with chromatin changes, enhanced ATM phosphorylation and prolonged G2/M cell cycle arrest. Our findings identify a β1 integrin/JNK co-dependent bypass signaling for GBM therapy resistance, which might be therapeutically exploitable.
Collapse
|
31
|
Klapproth E, Dickreuter E, Zakrzewski F, Seifert M, Petzold A, Dahl A, Schröck E, Klink B, Cordes N. Whole exome sequencing identifies mTOR and KEAP1 as potential targets for radiosensitization of HNSCC cells refractory to EGFR and β1 integrin inhibition. Oncotarget 2018; 9:18099-18114. [PMID: 29719593 PMCID: PMC5915060 DOI: 10.18632/oncotarget.24266] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/09/2018] [Indexed: 12/26/2022] Open
Abstract
Intrinsic and acquired resistances are major obstacles in cancer therapy. Genetic characterization is commonly used to identify predictive or prognostic biomarker signatures and potential cancer targets in samples from therapy-naïve patients. By far less common are such investigations to identify specific, predictive and/or prognostic gene signatures in patients or cancer cells refractory to a specific molecular-targeted intervention. This, however, might have a great value to foster the development of tailored, personalized cancer therapy. Based on our identification of a differential radiosensitization by single and combined β1 integrin (AIIB2) and EGFR (Cetuximab) targeting in more physiological, three-dimensional head and neck squamous cell carcinoma (HNSCC) cell cultures, we performed comparative whole exome sequencing, phosphoproteome analyses and RNAi knockdown screens in responder and non-responder cell lines. We found a higher rate of gene mutations with putative protein-changing characteristics in non-responders and different mutational profiles of responders and non-responders. These profiles allow stratification of HNSCC patients and identification of potential targets to address treatment resistance. Consecutively, pharmacological inhibition of mTOR and KEAP1 effectively diminished non-responder insusceptibility to β1 integrin and EGFR targeting for radiosensitization. Our data pinpoint the added value of genetic biomarker identification after selection for cancer subgroup responsiveness to targeted therapies.
Collapse
Affiliation(s)
- Erik Klapproth
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ellen Dickreuter
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
| | - Falk Zakrzewski
- German Cancer Consortium (DKTK), Dresden 01307, Germany
- German Cancer Research Center (DKFZ), Dresden partner site, Heidelberg 69120, Germany
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Dresden 01307, Germany
| | - Michael Seifert
- Institute for Medical Informatics and Biometry (IMB), Technische Universität Dresden, Dresden 01307, Germany
- National Center for Tumor Diseases (NCT), Dresden 01307, Germany
| | - Andreas Petzold
- Deep Sequencing Group, BIOTEChnology Center, Technische Universität Dresden, Dresden 01307, Germany
| | - Andreas Dahl
- Deep Sequencing Group, BIOTEChnology Center, Technische Universität Dresden, Dresden 01307, Germany
| | - Evelin Schröck
- German Cancer Consortium (DKTK), Dresden 01307, Germany
- German Cancer Research Center (DKFZ), Dresden partner site, Heidelberg 69120, Germany
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Dresden 01307, Germany
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Barbara Klink
- German Cancer Consortium (DKTK), Dresden 01307, Germany
- German Cancer Research Center (DKFZ), Dresden partner site, Heidelberg 69120, Germany
- Core Unit for Molecular Tumor Diagnostics (CMTD), National Center for Tumor Diseases (NCT), Dresden 01307, Germany
- Deep Sequencing Group, BIOTEChnology Center, Technische Universität Dresden, Dresden 01307, Germany
- Institute for Clinical Genetics, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Dresden 01307, Germany
- German Cancer Research Center (DKFZ), Dresden partner site, Heidelberg 69120, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology, Dresden 01328, Germany
| |
Collapse
|
32
|
Förster S, Hehlgans S, Rödel F, Otto B, Cordes N. Differential effects of α-catenin on the invasion and radiochemosensitivity of human colorectal cancer cells. Int J Oncol 2018; 52:1117-1128. [PMID: 29484367 PMCID: PMC5843400 DOI: 10.3892/ijo.2018.4279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 01/31/2018] [Indexed: 11/26/2022] Open
Abstract
Driven by genetic and epigenetic alterations, progression, therapy resistance and metastasis are frequent events in colorectal cancer (CRC). Although often speculated, the function of cell-cell contact for radiochemosensitivity, particularly associated with E-cadherin/catenin complex, warrants further clarification. In this study, we investigated the role of the E-cadherin/catenin complex proteins under more physiological three-dimensional (3D) cell culture conditions in a panel of CRC cell lines. In contrast to floating spheroids and growth in the laminin-rich matrix, collagen type 1 induced the formation of two distinct growth phenotypes, i.e., cell groups and single cells, in 5 out of the 8 CRC cell lines. Further characterization of these subpopulations revealed that, intriguingly, cell-cell contact proteins are important for invasion, but negligible for radiochemosensitivity, proliferation and adhesion. Despite the generation of genomic and transcriptomic data, we were unable to elucidate the mechanisms through which α-catenin affects collagen type 1 invasion. In a retrospective analysis of patients with rectal carcinoma, a low α-catenin expression trended with overall survival, as well as locoregional and distant control. Our results suggest that the E-cadherin/catenin complex proteins forming cell-cell contacts are mainly involved in the invasion, rather than the radiochemosensitivity of 3D grown CRC cells. Further studies are warranted in order to provide a better understanding of the molecular mechanisms controlling cell-cell adhesion in the context of radiochemoresistance.
Collapse
Affiliation(s)
- Sarah Förster
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, 01328 Dresden, Germany
| | - Stephanie Hehlgans
- Department of Radiotherapy and Oncology, University of Frankfurt, 60590 Frankfurt, Germany
| | - Franz Rödel
- Department of Radiotherapy and Oncology, University of Frankfurt, 60590 Frankfurt, Germany
| | | | - Nils Cordes
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, 01328 Dresden, Germany
| |
Collapse
|
33
|
Hohmann T, Grabiec U, Vogel C, Ghadban C, Ensminger S, Bache M, Vordermark D, Dehghani F. The Impact of Non-Lethal Single-Dose Radiation on Tumor Invasion and Cytoskeletal Properties. Int J Mol Sci 2017; 18:E2001. [PMID: 28926987 PMCID: PMC5618650 DOI: 10.3390/ijms18092001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 12/27/2022] Open
Abstract
Irradiation is the standard therapy for glioblastoma multiforme. Glioblastoma are highly resistant to radiotherapy and the underlying mechanisms remain unclear. To better understand the biological effects of irradiation on glioblastoma cells, we tested whether nonlethal irradiation influences the invasiveness, cell stiffness, and actin cytoskeleton properties. Two different glioblastoma cell lines were irradiated with 2 Gy and changes in mechanical and migratory properties and alterations in the actin structure were measured. The invasiveness of cell lines was determined using a co-culture model with organotypic hippocampal slice cultures. Irradiation led to changes in motility and a less invasive phenotype in both investigated cell lines that were associated with an increase in a "generalized stiffness" and changes in the actin structure. In this study we demonstrate that irradiation can induce changes in the actin cytoskeleton and motility, which probably results in reduced invasiveness of glioblastoma cell lines. Furthermore, "generalized stiffness" was shown to be a profound marker of the invasiveness of a tumor cell population in our model.
Collapse
Affiliation(s)
- Tim Hohmann
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Urszula Grabiec
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Carolin Vogel
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Chalid Ghadban
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| | - Stephan Ensminger
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Matthias Bache
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Dirk Vordermark
- Department of Radiation Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120 Halle, Germany.
| | - Faramarz Dehghani
- Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, 06108 Halle, Germany.
| |
Collapse
|
34
|
Dickreuter E, Cordes N. The cancer cell adhesion resistome: mechanisms, targeting and translational approaches. Biol Chem 2017; 398:721-735. [PMID: 28002024 DOI: 10.1515/hsz-2016-0326] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
Abstract
Cell adhesion-mediated resistance limits the success of cancer therapies and is a great obstacle to overcome in the clinic. Since the 1990s, where it became clear that adhesion of tumor cells to the extracellular matrix is an important mediator of therapy resistance, a lot of work has been conducted to understand the fundamental underlying mechanisms and two paradigms were deduced: cell adhesion-mediated radioresistance (CAM-RR) and cell adhesion-mediated drug resistance (CAM-DR). Preclinical work has evidently demonstrated that targeting of integrins, adapter proteins and associated kinases comprising the cell adhesion resistome is a promising strategy to sensitize cancer cells to both radiotherapy and chemotherapy. Moreover, the cell adhesion resistome fundamentally contributes to adaptation mechanisms induced by radiochemotherapy as well as molecular drugs to secure a balanced homeostasis of cancer cells for survival and growth. Intriguingly, this phenomenon provides a basis for synthetic lethal targeted therapies simultaneously administered to standard radiochemotherapy. In this review, we summarize current knowledge about the cell adhesion resistome and highlight targeting strategies to override CAM-RR and CAM-DR.
Collapse
Affiliation(s)
| | - Nils Cordes
- , Faculty of Medicine and University Hospital Carl Gustav Carus
| |
Collapse
|
35
|
Cihoric N, Tsikkinis A, Minniti G, Lagerwaard FJ, Herrlinger U, Mathier E, Soldatovic I, Jeremic B, Ghadjar P, Elicin O, Lössl K, Aebersold DM, Belka C, Herrmann E, Niyazi M. Current status and perspectives of interventional clinical trials for glioblastoma - analysis of ClinicalTrials.gov. Radiat Oncol 2017; 12:1. [PMID: 28049492 PMCID: PMC5210306 DOI: 10.1186/s13014-016-0740-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/08/2016] [Indexed: 01/09/2023] Open
Abstract
The records of 208.777 (100%) clinical trials registered at ClinicalTrials.gov were downloaded on the 19th of February 2016. Phase II and III trials including patients with glioblastoma were selected for further classification and analysis. Based on the disease settings, trials were classified into three groups: newly diagnosed glioblastoma, recurrent disease and trials with no differentiation according to disease setting. Furthermore, we categorized trials according to the experimental interventions, the primary sponsor, the source of financial support and trial design elements. Trends were evaluated using the autoregressive integrated moving average model. Two hundred sixteen (0.1%) trials were selected for further analysis. Academic centers (investigator initiated trials) were recorded as primary sponsors in 56.9% of trials, followed by industry 25.9%. Industry was the leading source of monetary support for the selected trials in 44.4%, followed by 25% of trials with primarily academic financial support. The number of newly initiated trials between 2005 and 2015 shows a positive trend, mainly through an increase in phase II trials, whereas phase III trials show a negative trend. The vast majority of trials evaluate forms of different systemic treatments (91.2%). In total, one hundred different molecular entities or biologicals were identified. Of those, 60% were involving drugs specifically designed for central nervous system malignancies. Trials that specifically address radiotherapy, surgery, imaging and other therapeutic or diagnostic methods appear to be rare. Current research in glioblastoma is mainly driven or sponsored by industry, academic medical oncologists and neuro-oncologists, with the majority of trials evaluating forms of systemic therapies. Few trials reach phase III. Imaging, radiation therapy and surgical procedures are underrepresented in current trials portfolios. Optimization in research portfolio for glioblastoma is needed.
Collapse
Affiliation(s)
- Nikola Cihoric
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland.
| | - Alexandros Tsikkinis
- Department of Genitourinary Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Giuseppe Minniti
- Unit of Radiation Oncology, Sant' Andrea Hospital, University Sapienza, and IRCCS Neuromed, Pozzilli (IS), Italy
| | - Frank J Lagerwaard
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Ulrich Herrlinger
- Department of Neurology, Division of Clinical Neurooncology, University of Bonn Medical Center, Bonn, Germany
| | - Etienne Mathier
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Ivan Soldatovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Branislav Jeremic
- Institute of Lung Diseases, Sremska Kamenica, Serbia and BioIRC Center for Biomedical Research, Kragujevac, Serbia
| | - Pirus Ghadjar
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Olgun Elicin
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Kristina Lössl
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Daniel M Aebersold
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Claus Belka
- Department of Radiation Oncology, LMU Munich, München, Germany.,German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Evelyn Herrmann
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, 3010, Bern, Switzerland
| | - Maximilian Niyazi
- Department of Radiation Oncology, LMU Munich, München, Germany.,German Cancer Consortium (DKTK) & German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
36
|
Panzetta V, De Menna M, Musella I, Pugliese M, Quarto M, Netti PA, Fusco S. X-rays effects on cytoskeleton mechanics of healthy and tumor cells. Cytoskeleton (Hoboken) 2016; 74:40-52. [DOI: 10.1002/cm.21334] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/09/2016] [Accepted: 08/15/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Valeria Panzetta
- Center for Advanced Biomaterials for Health Care@CRIB - Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci n. 53 Napoli 80125 Italy
| | - Marta De Menna
- Department of Experimental and Clinic Medicine; University of Catanzaro Magna Graecia; Catanzaro Italy
| | - Ida Musella
- Center for Advanced Biomaterials for Health Care@CRIB - Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci n. 53 Napoli 80125 Italy
| | - Mariagabriella Pugliese
- Dipartimento di Fisica; Università Federico II and INFN-Sezione di Napoli; Monte S. Angelo, Via Cintia Napoli 80126 Italy
| | - Maria Quarto
- Dipartimento di Fisica; Università Federico II and INFN-Sezione di Napoli; Monte S. Angelo, Via Cintia Napoli 80126 Italy
| | - Paolo A. Netti
- Center for Advanced Biomaterials for Health Care@CRIB - Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci n. 53 Napoli 80125 Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II; P.le Tecchio 80 Napoli 80125 Italy
| | - Sabato Fusco
- Center for Advanced Biomaterials for Health Care@CRIB - Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci n. 53 Napoli 80125 Italy
| |
Collapse
|
37
|
Hausmann C, Temme A, Cordes N, Eke I. ILKAP, ILK and PINCH1 control cell survival of p53-wildtype glioblastoma cells after irradiation. Oncotarget 2016; 6:34592-605. [PMID: 26460618 PMCID: PMC4741475 DOI: 10.18632/oncotarget.5423] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/25/2015] [Indexed: 11/29/2022] Open
Abstract
The prognosis is generally poor for patients suffering from glioblastoma multiforme (GBM) due to radiation and drug resistance. Prosurvival signaling originating from focal adhesion hubs essentially contributes to therapy resistance and tumor aggressiveness. As the underlying molecular mechanisms remain largely elusive, we addressed whether targeting of the focal adhesion proteins particularly interesting new cysteine-histidine-rich 1 (PINCH1), integrin-linked kinase (ILK) and ILK associated phosphatase (ILKAP) modulates GBM cell radioresistance. Intriguingly, PINCH1, ILK and ILKAP depletion sensitized p53-wildtype, but not p53-mutant, GBM cells to radiotherapy. Concomitantly, these cells showed inactivated Glycogen synthase kinase-3β (GSK3β) and reduced proliferation. For PINCH1 and ILKAP knockdown, elevated levels of radiation-induced γH2AX/53BP1-positive foci, as a marker for DNA double strand breaks, were observed. Mechanistically, we identified radiation-induced phosphorylation of DNA protein kinase (DNAPK), an important DNA repair protein, to be dependent on ILKAP. This interaction was fundamental to radiation survival of p53-wildtype GBM cells. Conclusively, our data suggest an essential role of PINCH1, ILK and ILKAP for the radioresistance of p53-wildtype GBM cells and provide evidence for DNAPK functioning as a central mediator of ILKAP signaling. Strategies for targeting focal adhesion proteins in combination with radiotherapy might be a promising approach for patients with GBM.
Collapse
Affiliation(s)
- Christina Hausmann
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Achim Temme
- Section of Experimental Neurosurgery/Tumor Immunology, Department of Neurosurgery University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Department of Radiation Oncology, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology, 01328 Dresden, Germany.,German Cancer Consortium (DKTK), 01307 Dresden, Germany.,German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Iris Eke
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.,Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health/National Cancer Institute, Bethesda, MD 20892, USA
| |
Collapse
|
38
|
Walenta S, Mueller-Klieser W. Differential Superiority of Heavy Charged-Particle Irradiation to X-Rays: Studies on Biological Effectiveness and Side Effect Mechanisms in Multicellular Tumor and Normal Tissue Models. Front Oncol 2016; 6:30. [PMID: 26942125 PMCID: PMC4766872 DOI: 10.3389/fonc.2016.00030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/28/2016] [Indexed: 01/31/2023] Open
Abstract
This review is focused on the radiobiology of carbon ions compared to X-rays using multicellular models of tumors and normal mucosa. The first part summarizes basic radiobiological effects, as observed in cancer cells. The second, more clinically oriented part of the review, deals with radiation-induced cell migration and mucositis. Multicellular spheroids from V79 hamster cells were irradiated with X-rays or carbon ions under ambient or restricted oxygen supply conditions. Reliable oxygen enhancement ratios could be derived to be 2.9, 2.8, and 1.4 for irradiation with photons, 12C+6 in the plateau region, and 12C+6 in the Bragg peak, respectively. Similarly, a relative biological effectiveness of 4.3 and 2.1 for ambient pO2 and hypoxia was obtained, respectively. The high effectiveness of carbon ions was reflected by an enhanced accumulation of cells in G2/M and a dose-dependent massive induction of apoptosis. These data clearly show that heavy charged particles are more efficient in sterilizing tumor cells than conventional irradiation even under hypoxic conditions. Clinically relevant doses (3 Gy) of X-rays induced an increase in migratory activity of U87 but not of LN229 or HCT116 tumor cells. Such an increase in cell motility following irradiation in situ could be the source of recurrence. In contrast, carbon ion treatment was associated with a dose-dependent decrease in migration with all cell lines and under all conditions investigated. The radiation-induced loss of cell motility was correlated, in most cases, with corresponding changes in β1 integrin expression. The photon-induced increase in cell migration was paralleled by an elevated phosphorylation status of the epidermal growth factor receptor and AKT-ERK1/2 pathway. Such a hyperphosphorylation did not occur during 12C+6 irradiation under all conditions registered. Comparing the gene toxicity of X-rays with that of particles using the γH2AX technique in organotypic cultures of the oral mucosa, the superior effectiveness of heavy ions was confirmed by a twofold higher number of foci per nucleus. However, proinflammatory signs were similar for both treatment modalities, e.g., the activation of NFκB and the release of IL6 and IL8. The presence of peripheral blood mononuclear cell increased the radiation-induced release of the proinflammatory cytokines by factors of 2–3. Carbon ions are part of the cosmic radiation. Long-term exposure to such particles during extended space flights, as planned by international space agencies, may thus impose a medical and safety risk on the astronauts by a potential induction of mucositis. In summary, particle irradiation is superior to gamma-rays due to a higher radiobiological effectiveness, a reduced hypoxia-induced radioresistance, a multicellular radiosensitization, and the absence of a radiation-induced cell motility. However, the potential of inducing mucositis is similar for both radiation types.
Collapse
Affiliation(s)
- Stefan Walenta
- Institute of Pathophysiology, University Medical Center, University of Mainz , Mainz , Germany
| | | |
Collapse
|
39
|
Glioblastoma Stem Cells Microenvironment: The Paracrine Roles of the Niche in Drug and Radioresistance. Stem Cells Int 2016; 2016:6809105. [PMID: 26880981 PMCID: PMC4736577 DOI: 10.1155/2016/6809105] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 11/09/2015] [Accepted: 11/10/2015] [Indexed: 12/13/2022] Open
Abstract
Among all solid tumors, the high-grade glioma appears to be the most vascularized one. In fact, "microvascular hyperplasia" is a hallmark of GBM. An altered vascular network determines irregular blood flow, so that tumor cells spread rapidly beyond the diffusion distance of oxygen in the tissue, with the consequent formation of hypoxic or anoxic areas, where the bulk of glioblastoma stem cells (GSCs) reside. The response to this event is the induction of angiogenesis, a process mediated by hypoxia inducible factors. However, this new capillary network is not efficient in maintaining a proper oxygen supply to the tumor mass, thereby causing an oxygen gradient within the neoplastic zone. This microenvironment helps GSCs to remain in a "quiescent" state preserving their potential to proliferate and differentiate, thus protecting them by the effects of chemo- and radiotherapy. Recent evidences suggest that responses of glioblastoma to standard therapies are determined by the microenvironment of the niche, where the GSCs reside, allowing a variety of mechanisms that contribute to the chemo- and radioresistance, by preserving GSCs. It is, therefore, crucial to investigate the components/factors of the niche in order to formulate new adjuvant therapies rendering more efficiently the gold standard therapies for this neoplasm.
Collapse
|
40
|
Vehlow A, Storch K, Matzke D, Cordes N. Molecular Targeting of Integrins and Integrin-Associated Signaling Networks in Radiation Oncology. Recent Results Cancer Res 2016; 198:89-106. [PMID: 27318682 DOI: 10.1007/978-3-662-49651-0_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Radiation and chemotherapy are the main pillars of the current multimodal treatment concept for cancer patients. However, tumor recurrences and resistances still hamper treatment success regardless of advances in radiation beam application, particle radiotherapy, and optimized chemotherapeutics. To specifically intervene at key recurrence- and resistance-promoting molecular processes, the development of potent and specific molecular-targeted agents is demanded for an efficient, safe, and simultaneous integration into current standard of care regimens. Potential targets for such an approach are integrins conferring structural and biochemical communication between cells and their microenvironment. Integrin binding to extracellular matrix activates intracellular signaling for regulating essential cellular functions such as survival, proliferation, differentiation, adhesion, and cell motility. Tumor-associated characteristics such as invasion, metastasis, and radiochemoresistance also highly depend on integrin function. Owing to their dual functionality and their overexpression in the majority of human malignancies, integrins present ideal and accessible targets for cancer therapy. In the following chapter, the current knowledge on aspects of the tumor microenvironment, the molecular regulation of integrin-dependent radiochemoresistance and current approaches to integrin targeting are summarized.
Collapse
Affiliation(s)
- Anne Vehlow
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Katja Storch
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Daniela Matzke
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nils Cordes
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
| |
Collapse
|
41
|
Chen L, Kang C. miRNA interventions serve as 'magic bullets' in the reversal of glioblastoma hallmarks. Oncotarget 2015; 6:38628-42. [PMID: 26439688 PMCID: PMC4770725 DOI: 10.18632/oncotarget.5926] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 09/06/2015] [Indexed: 01/04/2023] Open
Abstract
microRNAs (miRNAs) are no longer deemed small pieces of RNA "trash" in the human transcriptome but are considered to be master regulators of gene expression that are critical in maintaining cellular homeostasis post-transcriptionally. The concept triggers great interest in studying miRNA dysregulations in human diseases, especially in cancers. Glioblastoma (GBM) has long been the leading cause of the high mortality and morbidity of CNS tumors in adults, which is a consequence of the lack of strategies to reverse the hallmark features of GBM (e.g., borderless expansion and diffuse infiltration). In the past decade, dissecting the molecular architecture of GBM has led to a better understanding of the molecular basis of the hallmarks, generating many promising pharmacological protein targets. However, few clinical responses have been highlighted, suggesting the demand for new therapeutic strategies and targets. In this review, we systemically summarize the context-dependently validated miRNAs with one or more functional targets in the development of GBM hallmarks and review the current miRNA-targeting strategies. We note that only a few miRNA-based therapeutics are trialed for clinical significance, and none of them is tailored to GBM, thereby urging us to bring miRNA therapeutics to the front line either alone or in combination.
Collapse
Affiliation(s)
- Luyue Chen
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunsheng Kang
- Laboratory of Neuro-Oncology, Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
42
|
Artacho-Cordón F, Ríos-Arrabal S, Olivares-Urbano MA, Storch K, Dickreuter E, Muñoz-Gámez JA, León J, Calvente I, Torné P, Salinas MDM, Cordes N, Núñez MI. Valproic acid modulates radiation-enhanced matrix metalloproteinase activity and invasion of breast cancer cells. Int J Radiat Biol 2015; 91:946-56. [PMID: 26490761 DOI: 10.3109/09553002.2015.1087067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE To evaluate matrix metalloproteinase (MMP) activity and invasion after ionizing radiation (IR) exposure and to determine whether MMP could be epigenetically modulated by histone deacetylase (HDAC) inhibition. MATERIAL AND METHODS Two human breast cancer cell lines (MDA-MB-231 and MCF-7) were cultured in monolayer (2D) and in laminin-rich extracellular matrix (3D). Invasion capability, collagenolytic and gelatinolytic activity, MMP and TIMP protein and mRNA expression and clonogenic survival were analyzed after IR exposure, with and without a HDAC inhibition treatment [1.5 mM valproic acid (VA) or 1 μM trichostatin-A (TSA)]. RESULTS IR exposure resulted in cell line-dependent stimulation of invasion capacity. In contrast to MCF-7 cells, irradiated MDA-MB-231 showed significantly enhanced mRNA expression of mmp-1, mmp-3 and mmp-13 and of their regulators timp-1 and timp-2 relative to unirradiated controls. This translated into increased collagenolytic and gelatinolytic activity and could be reduced after valproic acid (VA) treatment. Additionally, VA also mitigated IR-enhanced mmp and timp mRNA expression as well as IR-increased invasion capability. Finally, our data confirm the radiosensitizing effect of VA. CONCLUSION These results suggest that IR cell line-dependently induces upregulation of MMP mRNA expression, which appears to be mechanistically linked to a higher invasion capability that is modifiable by HDAC inhibition.
Collapse
Affiliation(s)
- Francisco Artacho-Cordón
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Sandra Ríos-Arrabal
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| | | | - Katja Storch
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - Ellen Dickreuter
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - José Antonio Muñoz-Gámez
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Josefa León
- b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,i CIBER on hepatic and digestive diseases (CIBEREHD) , Spain
| | - Irene Calvente
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain
| | - Pablo Torné
- j General Surgery Management Unit, San Cecilio University Hospital , Granada , Spain
| | - María del Mar Salinas
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain
| | - Nils Cordes
- d OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, and Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany.,e Department of Radiation Oncology , University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany.,f German Cancer Consortium (DKTK), Dresden, Germany.,g German Cancer Research Center (DKFZ) , Heidelberg , Germany.,h Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf , Dresden , Germany
| | - María Isabel Núñez
- a Department of Radiology and Physical Medicine , University of Granada , Granada , Spain.,b Biosanitary Institute of Granada (ibs.GRANADA), University Hospitals of Granada/University of Granada , Granada , Spain.,c Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada , Armilla, Granada , Spain
| |
Collapse
|
43
|
Sforna L, Cenciarini M, Belia S, D'Adamo MC, Pessia M, Franciolini F, Catacuzzeno L. The role of ion channels in the hypoxia-induced aggressiveness of glioblastoma. Front Cell Neurosci 2015; 8:467. [PMID: 25642170 PMCID: PMC4295544 DOI: 10.3389/fncel.2014.00467] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/24/2014] [Indexed: 12/16/2022] Open
Abstract
The malignancy of glioblastoma multiform (GBM), the most common and aggressive form of human brain tumors, strongly correlates with the presence of hypoxic areas, but the mechanisms controlling the hypoxia-induced aggressiveness are still unclear. GBM cells express a number of ion channels whose activity supports cell volume changes and increases in the cytosolic Ca2+ concentration, ultimately leading to cell proliferation, migration or death. In several cell types it has previously been shown that low oxygen levels regulate the expression and activity of these channels, and more recent data indicate that this also occurs in GBM cells. Based on these findings, it may be hypothesized that the modulation of ion channel activity or expression by the hypoxic environment may participate in the acquisition of the aggressive phenotype observed in GBM cells residing in a hypoxic environment. If this hypothesis will be confirmed, the use of available ion channels modulators may be considered for implementing novel therapeutic strategies against these tumors.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Marta Cenciarini
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Silvia Belia
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Maria Cristina D'Adamo
- Faculty of Medicine, Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Mauro Pessia
- Faculty of Medicine, Section of Physiology and Biochemistry, Department of Experimental Medicine, University of Perugia Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia Perugia, Italy
| |
Collapse
|
44
|
Zheng Q, Liu Y, Zhou HJ, Du YT, Zhang BP, Zhang J, Miao GY, Liu B, Zhang H. X-ray radiation promotes the metastatic potential of tongue squamous cell carcinoma cells via modulation of biomechanical and cytoskeletal properties. Hum Exp Toxicol 2015; 34:894-903. [PMID: 25586002 DOI: 10.1177/0960327114561664] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study investigated the metastatic potential of tongue squamous cell carcinoma (TSCC) cells after X-ray irradiation as well as radiation-induced changes in the biomechanical properties and cytoskeletal structure that are relevant to metastasis. Tca-8113 TSCC cells were X-ray-irradiated at increasing doses (0, 1, 2, or 4 Gy), and 24 h later, migration was evaluated with the wound healing and transwell migration assays, while invasion was assessed with the Matrigel invasion assay. Confocal and atomic force microscopy were used to examine changes in the structure of the actin cytoskeleton and Young's modulus (cell stiffness), respectively. X-ray radiation induced dose-dependent increases in invasive and migratory potentials of cells relative to unirradiated control cells (p < 0.05). The Young's modulus of irradiated cells was decreased by radiation exposure (p < 0.05), which was accompanied by alterations in the integrity and organization of the cytoskeletal network, as evidenced by a decrease in the signal intensity of actin fibers (p < 0.05). X-ray irradiation enhanced migration and invasiveness in Tca-8113 TSCC cells by altering their biomechanical properties and the organization of the actin cytoskeleton. A biomechanics-based analysis can provide an additional platform for assessing tumor response to radiation and optimization of cancer therapies.
Collapse
Affiliation(s)
- Q Zheng
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - Y Liu
- Department of Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, People's Republic of China
| | - H J Zhou
- School of Stomatology, Northwest University for Nationalities, Lanzhou, Gansu, People's Republic of China
| | - Y T Du
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - B P Zhang
- School of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - J Zhang
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| | - G Y Miao
- Department of Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, People's Republic of China
| | - B Liu
- School of Stomatology, Lanzhou University, Lanzhou, Gansu, People's Republic of China Corresponding authors with equal contribution
| | - H Zhang
- Department of Radiation Biology and Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, Gansu, People's Republic of China Corresponding authors with equal contribution
| |
Collapse
|
45
|
Broustas CG, Lieberman HB. RAD9 enhances radioresistance of human prostate cancer cells through regulation of ITGB1 protein levels. Prostate 2014; 74:1359-70. [PMID: 25111005 PMCID: PMC4142073 DOI: 10.1002/pros.22842] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/03/2014] [Indexed: 01/17/2023]
Abstract
BACKGROUND Mouse embryonic stem cells null for Rad9 are sensitive to deleterious effects of ionizing radiation exposure. Likewise, integrin β1 is a known radioprotective factor. Previously, we showed that RAD9 downregulation in human prostate cancer cells reduces integrin β1 protein levels and ectopic expression of Mrad9 restores inherent high levels. METHODS We used RNA interference to knockdown Rad9 expression in PC3 and DU145 prostate cancer cells. These cells were then exposed to ionizing radiation, and integrin β1 protein levels were measured by immunoblotting. Survival of irradiated cells was measured by clonogenicity, cell cycle analysis, PARP-1 cleavage, and trypan blue exclusion. RESULTS The function of RAD9 in controlling integrin β1 expression is unique and not shared by the other members of the 9-1-1 complex, HUS1 and RAD1. RAD9 or integrin β1 silencing sensitizes DU145 and PC3 cells to ionizing radiation. Irradiation of DU145 cells with low levels of RAD9 induces cleavage of PARP-1 protein. High levels of ionizing radiation have no effect on integrin β1 protein levels. However, when RAD9 downregulation is combined with 10 Gy of ionizing radiation in DU145 or PC3 cells, there is an additional 50% downregulation of integrin β1 compared with levels in unirradiated RAD9 knockdown cells. Finally, PC3 cells growing on fibronectin display increased radioresistance. However, PC3 cells with RAD9 knockdown are no longer protected by fibronectin after treatment with ionizing radiation. CONCLUSIONS Downregulation of RAD9 when combined with ionizing radiation results in reduction of ITGB1 protein levels in prostate cancer cells, and increased lethality.
Collapse
Affiliation(s)
- Constantinos G. Broustas
- Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, NY 10032
| | - Howard B. Lieberman
- Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, NY 10032
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032
| |
Collapse
|
46
|
Han N, Shahveranov A, Cheng Y, Qin K, Yu SY, Zhang MX. Effects of connective tissue growth factor (CTGF) gene silencing on the radiosensitivity of glioblastoma. Int J Clin Exp Med 2014; 7:2557-63. [PMID: 25356109 PMCID: PMC4211759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 08/16/2014] [Indexed: 06/04/2023]
Abstract
The effects of connective tissue growth factor (CTGF) gene silencing on the radiosensitivity of glioblastoma cells (GBM) were investigated. The lentivirus-mediated short hairpin RNA (shRNA) expression vector targeting CTGF was constructed and transinfected into U87MG human GBM cell line. The CTGF gene expression in U87MG cells was significantly down-regulated. After irradiation with 6 MV X-rays at a dose rate of 2.5 Gy/min, the clonogenicity, proliferation and migration of U87MG cells were assayed in vitro. The survival, proliferation and migration of U87MG cells were all remarkably inhibited by CTGF silencing (p < 0.05 vs control). Our results demonstrate that CTGF is important for GBM and CTGF gene silencing can be a potential tool to enhance the sensitivity of GBM to radiotherapy.
Collapse
Affiliation(s)
- Na Han
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| | - Allahverdi Shahveranov
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| | - Yi Cheng
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| | - Kai Qin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| | - Shi-Ying Yu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| | - Meng-Xian Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030, China
| |
Collapse
|
47
|
Eke I, Cordes N. Focal adhesion signaling and therapy resistance in cancer. Semin Cancer Biol 2014; 31:65-75. [PMID: 25117005 DOI: 10.1016/j.semcancer.2014.07.009] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 12/18/2022]
Abstract
Interlocking gene mutations, epigenetic alterations and microenvironmental features perpetuate tumor development, growth, infiltration and spread. Consequently, intrinsic and acquired therapy resistance arises and presents one of the major goals to solve in oncologic research today. Among the myriad of microenvironmental factors impacting on cancer cell resistance, cell adhesion to the extracellular matrix (ECM) has recently been identified as key determinant. Despite the differentiation between cell adhesion-mediated drug resistance (CAMDR) and cell adhesion-mediated radioresistance (CAMRR), the underlying mechanisms share great overlap in integrin and focal adhesion hub signaling and differ further downstream in the complexity of signaling networks between tumor entities. Intriguingly, cell adhesion to ECM is per se also essential for cancer cells similar to their normal counterparts. However, based on the overexpression of focal adhesion hub signaling receptors and proteins and a distinct addiction to particular integrin receptors, targeting of focal adhesion proteins has been shown to potently sensitize cancer cells to different treatment regimes including radiotherapy, chemotherapy and novel molecular therapeutics. In this review, we will give insight into the role of integrins in carcinogenesis, tumor progression and metastasis. Additionally, literature and data about the function of focal adhesion molecules including integrins, integrin-associated proteins and growth factor receptors in tumor cell resistance to radio- and chemotherapy will be elucidated and discussed.
Collapse
Affiliation(s)
- Iris Eke
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Dresden 01328, Germany; Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Nils Cordes
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Dresden 01328, Germany; Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Radiation Oncology, Helmholtz-Zentrum Dresden - Rossendorf, Dresden 01328, Germany.
| |
Collapse
|
48
|
Orth M, Lauber K, Niyazi M, Friedl AA, Li M, Maihöfer C, Schüttrumpf L, Ernst A, Niemöller OM, Belka C. Current concepts in clinical radiation oncology. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2014; 53:1-29. [PMID: 24141602 PMCID: PMC3935099 DOI: 10.1007/s00411-013-0497-2] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/05/2013] [Indexed: 05/04/2023]
Abstract
Based on its potent capacity to induce tumor cell death and to abrogate clonogenic survival, radiotherapy is a key part of multimodal cancer treatment approaches. Numerous clinical trials have documented the clear correlation between improved local control and increased overall survival. However, despite all progress, the efficacy of radiation-based treatment approaches is still limited by different technological, biological, and clinical constraints. In principle, the following major issues can be distinguished: (1) The intrinsic radiation resistance of several tumors is higher than that of the surrounding normal tissue, (2) the true patho-anatomical borders of tumors or areas at risk are not perfectly identifiable, (3) the treatment volume cannot be adjusted properly during a given treatment series, and (4) the individual heterogeneity in terms of tumor and normal tissue responses toward irradiation is immense. At present, research efforts in radiation oncology follow three major tracks, in order to address these limitations: (1) implementation of molecularly targeted agents and 'omics'-based screening and stratification procedures, (2) improvement of treatment planning, imaging, and accuracy of dose application, and (3) clinical implementation of other types of radiation, including protons and heavy ions. Several of these strategies have already revealed promising improvements with regard to clinical outcome. Nevertheless, many open questions remain with individualization of treatment approaches being a key problem. In the present review, the current status of radiation-based cancer treatment with particular focus on novel aspects and developments that will influence the field of radiation oncology in the near future is summarized and discussed.
Collapse
Affiliation(s)
- Michael Orth
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maximilian Niyazi
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Anna A. Friedl
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Minglun Li
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Cornelius Maihöfer
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Lars Schüttrumpf
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Anne Ernst
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Olivier M. Niemöller
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
- Present Address: Clinic for Radiation Oncology, St. Elisabeth Hospital Ravensburg, Ravensburg, Germany
| | - Claus Belka
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
49
|
Vehlow A, Cordes N. Invasion as target for therapy of glioblastoma multiforme. Biochim Biophys Acta Rev Cancer 2013; 1836:236-44. [PMID: 23891970 DOI: 10.1016/j.bbcan.2013.07.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/09/2013] [Accepted: 07/18/2013] [Indexed: 12/27/2022]
Abstract
The survival of cancer patients suffering from glioblastoma multiforme is limited to just a few months even after treatment with the most advanced techniques. The indefinable borders of glioblastoma cell infiltration into the surrounding healthy tissue prevent complete surgical removal. In addition, genetic mutations, epigenetic modifications and microenvironmental heterogeneity cause resistance to radio- and chemotherapy altogether resulting in a hardly to overcome therapeutic scenario. Therefore, the development of efficient therapeutic strategies to combat these tumors requires a better knowledge of genetic and proteomic alterations as well as the infiltrative behavior of glioblastoma cells and how this can be targeted. Among many cell surface receptors, members of the integrin family are known to regulate glioblastoma cell invasion in concert with extracellular matrix degrading proteases. While preclinical and early clinical trials suggested specific integrin targeting as a promising therapeutic approach, clinical trials failed to deliver improved cure rates up to now. Little is known about glioblastoma cell motility, but switches in invasion modes and adaption to specific microenvironmental cues as a consequence of treatment may maintain tumor cell resistance to therapy. Thus, understanding the molecular basis of integrin and protease function for glioblastoma cell invasion in the context of radiochemotherapy is a pressing issue and may be beneficial for the design of efficient therapeutic approaches. This review article summarizes the latest findings on integrins and extracellular matrix in glioblastoma and adds some perspective thoughts on how this knowledge might be exploited for optimized multimodal therapy approaches.
Collapse
Affiliation(s)
- Anne Vehlow
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, 01307 Dresden, Germany
| | | |
Collapse
|
50
|
Stahler C, Roth J, Cordes N, Taucher-Scholz G, Mueller-Klieser W. Impact of carbon ion irradiation on epidermal growth factor receptor signaling and glioma cell migration in comparison to conventional photon irradiation. Int J Radiat Biol 2013; 89:454-61. [DOI: 10.3109/09553002.2013.766769] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|