1
|
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
|
2
|
Koosha F, Ahmadikamalabadi M, Mohammadi M. Review of Recent Improvements in Carbon Ion Radiation Therapy in the Treatment of Glioblastoma. Adv Radiat Oncol 2024; 9:101465. [PMID: 38770179 PMCID: PMC11103612 DOI: 10.1016/j.adro.2024.101465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/11/2024] [Indexed: 05/22/2024] Open
Abstract
Purpose This article provides an overview of the physical and biologic properties of carbon ions, followed by an examination of the latest clinical outcomes in patients with glioma who have received carbon ion radiation therapy. Methods and Materials According to thee articles that have been reviewed, glioma represents the predominant form of neoplastic growth in the brain, accounting for approximately 51% of all malignancies affecting the nervous system. Currently, high-grade glioma, specifically glioblastoma, comprises 15% of cases and is associated with a high mortality rate. The development of novel drugs for the treatment of high-grade tumors has been impeded by various factors, such as the blood-brain barrier and tumor heterogeneity, despite numerous endeavors. According to the definition of tumor grade established by the World Health Organization, the conventional treatment involves surgical resection followed by adjuvant radiation and chemotherapy. Despite numerous attempts in photon radiation therapy to apply the highest possible dose to the tumor site while minimizing damage to healthy tissue, there has been no success in increasing patient survival. The primary cause of resistance to conventional radiation therapy methods, namely x-ray and gamma-ray, is attributed to the survival of radio-resistant glioma stem cells, which have the potential to trigger a recurrence of tumors. Particle beams, such as protons and carbon ions, can deposit the highest dose to a confined region, thus offering a more accurate dose distribution compared with photon beams. Results Carbon ions exhibit higher linear energy transfer and relative biologic effectiveness compared with photons, potentially enabling them to overcome radio-resistant tumor cells. Conclusions Therefore, it can be hypothesized that carbon ion radiation therapy may show superior efficacy in destroying neoplastic cells with reduced negative outcomes compared with x-ray radiation therapy.
Collapse
Affiliation(s)
- Fereshteh Koosha
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Ahmadikamalabadi
- Social Determinants of Health Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Radiology Department, School of Paramedical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohadesseh Mohammadi
- Department of Radiology Technology, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Xu R, Yin P, Wei J, Ding Q. The role of matrix stiffness in breast cancer progression: a review. Front Oncol 2023; 13:1284926. [PMID: 37916166 PMCID: PMC10616305 DOI: 10.3389/fonc.2023.1284926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/04/2023] [Indexed: 11/03/2023] Open
Abstract
The significance of matrix stiffness in cancer development has been investigated in recent years. The gradual elastic force the extracellular matrix imparts to cells, known as matrix stiffness, is one of the most important types of mechanical stimulation. Increased matrix stiffness alters the biological activity of cells, which promotes the growth of numerous malignancies, including breast cancer. Comprehensive studies have demonstrated that increasing matrix stiffness activates molecular signaling pathways that are closely linked to breast cancer progression. There are many articles exploring the relationship between mechanism hardness and breast cancer, so we wanted to provide a systematic summary of recent research advances. In this review, we briefly introduce the mechanism of matrix stiffness in breast cancer, elaborate on the effect of extracellular matrix stiffness on breast cancer biological behavior and signaling pathways, and finally, we will talk about breast cancer treatment that focuses on matrix stiffness.
Collapse
Affiliation(s)
- Ruoxi Xu
- Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Peng Yin
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jifu Wei
- Department of Pharmacy, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Qiang Ding
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Vashishta M, Kumar V, Guha C, Wu X, Dwarakanath BS. Enhanced Glycolysis Confers Resistance Against Photon but Not Carbon Ion Irradiation in Human Glioma Cell Lines. Cancer Manag Res 2023; 15:1-16. [PMID: 36628255 PMCID: PMC9826608 DOI: 10.2147/cmar.s385968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/17/2022] [Indexed: 01/05/2023] Open
Abstract
Purpose Metabolic reprogramming is a key hallmark in various malignancies and poses a challenge in achieving success with various therapies. Enhanced glycolysis is known to confer resistance against photon irradiation while the tumor response to carbon ion irradiation (CII) has not been investigated. This study aimed to investigate the effects of enhanced glycolysis on the response of human glioma cell lines to CII compared to the response to X-rays. Material and Methods Glycolysis was stimulated using Dinitrophenol (DNP), a mild OXPHOS inhibitor, in three human glioma cell lines (U251, U87, and LN229) and assessed by monitoring glucose uptake and utilization as well as expression of regulators of glycolysis (glucose transporter protein type 1(Glut1), hexokinase-II (HKII), and Pyruvate Kinase-2 (PKM2). Radiation (X-rays and CII) induced loss of clonogenic survival growth inhibition and perturbations in cell cycle progression (G2+M block), cytogenetic damage (micronuclei formation), apoptosis, necrosis (reflecting interphase death), and cell migration (Scratch assay) were investigated as parameters of radiation response. Results DNP (1 mM) enhanced the expression levels of GLUT1, HKII, and PKM2 by 30-60% and glucose uptake as well as usage by nearly 3 folds in U251 cells suggesting the stimulation of glycolysis. Enhanced glycolysis attenuated the loss of clonogenic survival with D10 doses increasing by 20% to 65% in these cell lines, while no significant changes were noted following CII. Concomitantly, dose-dependent growth inhibition, and cytogenetic damage as well as apoptosis and necrosis induced by X-rays were also reduced by elevated glycolysis in U251 and LN229 cells by 20-50%. However, stimulation of glycolysis enhanced the X-ray-induced cell migration, while it had negligible effect on migration following CII. Conclusion Our results suggest that enhanced glycolysis confers resistance against X-ray-induced cell death and migration, while it may not significantly alter the cellular responses to carbon ion irradiation.
Collapse
Affiliation(s)
- Mohit Vashishta
- R&D Department, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, People’s Republic of China,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, People’s Republic of China,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, People’s Republic of China,Rangel College of Pharmacy, Texas A&M University, College Station, TX, USA
| | - Vivek Kumar
- R&D Department, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, People’s Republic of China,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, People’s Republic of China,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, People’s Republic of China
| | - Chandan Guha
- Albert Einstein College of Medicine, The Bronx, NY, USA
| | - Xiaodong Wu
- R&D Department, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, People’s Republic of China,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, People’s Republic of China,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, People’s Republic of China
| | - Bilikere S Dwarakanath
- R&D Department, Shanghai Proton and Heavy Ion Center (SPHIC), Shanghai, People’s Republic of China,Shanghai Key Laboratory of Radiation Oncology (20dz2261000), Shanghai, People’s Republic of China,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, People’s Republic of China,Central Research Facility, Sri Ramachandra Institute of Higher Education and Research, Porur, ChennaiIndia,Indian Academy Degree College Autonomous (IADC-A), Bengaluru, Karnataka, India,Correspondence: Bilikere S Dwarakanath, Indian Academy Degree College Autonomous (IADC-A), 230, Hennur Main Rd, Meganahalli, Kalyan Nagar, Bengaluru, Karnataka, 560043, India, Tel +91 9952081077, Email
| |
Collapse
|
5
|
Utilizing Carbon Ions to Treat Medulloblastomas that Exhibit Chromothripsis. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-022-00213-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Abstract
Purpose of Review
Novel radiation therapies with accelerated charged particles such as protons and carbon ions have shown encouraging results in oncology. We present recent applications as well as benefits and risks associated with their use.
Recent Findings
We discuss the use of carbon ion radiotherapy to treat a specific type of aggressive pediatric brain tumors, namely medulloblastomas with chromothripsis. Potential reasons for the resistance to conventional treatment, such as the presence of cancer stem cells with unique properties, are highlighted. Finally, advantages of particle radiation alone and in combination with other therapies to overcome resistance are featured.
Summary
Provided that future preclinical studies confirm the evidence of high effectiveness, favorable toxicity profiles, and no increased risk of secondary malignancy, carbon ion therapy may offer a promising tool in pediatric (neuro)oncology and beyond.
Collapse
|
6
|
Loap P, De Marzi L, Almeida CE, Barcellini A, Bradley J, de Santis MC, Dendale R, Jimenez R, Orlandi E, Kirova Y. Hadrontherapy techniques for breast cancer. Crit Rev Oncol Hematol 2021; 169:103574. [PMID: 34958916 DOI: 10.1016/j.critrevonc.2021.103574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/31/2022] Open
Abstract
Radiotherapy plays a key role in breast cancer treatment, and recent technical advances have been made to improve the therapeutic window by limiting the risk of radiation-induced toxicity or by increasing tumor control. Hadrontherapy is a form a radiotherapy relying on particle beams; compared with photon beams, particle beams have specific physical, radiobiological and immunological properties, which can be valuable in diverse clinical situations. To date, available hadrontherapy techniques for breast cancer irradiation include proton therapy, carbon ion radiation therapy, fast neutron therapy and boron neutron capture therapy. This review analyzes the current rationale and level of evidence for each hadrontherapy technique for breast cancer.
Collapse
Affiliation(s)
- Pierre Loap
- Proton Therapy Center, Institut Curie, Orsay, France.
| | | | - Carlos Eduardo Almeida
- Department of Radiological Sciences, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | - Julie Bradley
- University of Florida Health Proton Therapy Institute, Jacksonville, FL, United States
| | | | - Remi Dendale
- Proton Therapy Center, Institut Curie, Orsay, France
| | - Rachel Jimenez
- Massachusetts General Hospital, Boston, MA, United States
| | - Ester Orlandi
- National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Youlia Kirova
- Proton Therapy Center, Institut Curie, Orsay, France
| |
Collapse
|
7
|
Loap P, Vischioni B, Bonora M, Ingargiola R, Ronchi S, Vitolo V, Barcellini A, Goanta L, De Marzi L, Dendale R, Pacelli R, Locati L, Calugaru V, Mammar H, Cavalieri S, Kirova Y, Orlandi E. Biological Rationale and Clinical Evidence of Carbon Ion Radiation Therapy for Adenoid Cystic Carcinoma: A Narrative Review. Front Oncol 2021; 11:789079. [PMID: 34917512 PMCID: PMC8668942 DOI: 10.3389/fonc.2021.789079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 12/13/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is a rare, basaloid, epithelial tumor, arising mostly from salivary glands. Radiation therapy can be employed as a single modality for unresectable tumors, in an adjuvant setting after uncomplete resection, in case of high-risk pathological features, or for recurrent tumors. Due to ACC intrinsic radioresistance, high linear energy transfer (LET) radiotherapy techniques have been evaluated for ACC irradiation: while fast neutron therapy has now been abandoned due to toxicity concerns, charged particle beams such as protons and carbon ions are at present the beams used for hadron therapy. Carbon ion radiation therapy (CIRT) is currently increasingly used for ACC irradiation. The aim of this review is to describe the immunological, molecular and clinicopathological bases that support ACC treatment with CIRT, as well as to expose the current clinical evidence that reveal the advantages of using CIRT for treating ACC.
Collapse
Affiliation(s)
- Pierre Loap
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy.,Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Barbara Vischioni
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Maria Bonora
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Rossana Ingargiola
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Sara Ronchi
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Viviana Vitolo
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Amelia Barcellini
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| | - Lucia Goanta
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Ludovic De Marzi
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France.,Institut Curie, PSL Research University, University Paris Saclay, INSERM LITO, Orsay, France
| | - Remi Dendale
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Roberto Pacelli
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", Napoli, Italy
| | - Laura Locati
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valentin Calugaru
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Hamid Mammar
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Stefano Cavalieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Youlia Kirova
- Department of Radiation Oncology, Institut Curie, Paris, France.,Proton Therapy Center, Institut Curie, Orsay, France
| | - Ester Orlandi
- Radiation Oncology Unit, Clinical Department, National Center for Oncological Hadrontherapy (CNAO), Pavia, Italy
| |
Collapse
|
8
|
Xie Y, Han Y, Zhang X, Ma H, Li L, Yu R, Liu H. Application of New Radiosensitizer Based on Nano-Biotechnology in the Treatment of Glioma. Front Oncol 2021; 11:633827. [PMID: 33869019 PMCID: PMC8044949 DOI: 10.3389/fonc.2021.633827] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 03/03/2021] [Indexed: 12/26/2022] Open
Abstract
Glioma is the most common intracranial malignant tumor, and its specific pathogenesis has been unclear, which has always been an unresolved clinical problem due to the limited therapeutic window of glioma. As we all know, surgical resection, chemotherapy, and radiotherapy are the main treatment methods for glioma. With the development of clinical trials and traditional treatment techniques, radiotherapy for glioma has increasingly exposed defects in the treatment effect. In order to improve the bottleneck of radiotherapy for glioma, people have done a lot of work; among this, nano-radiosensitizers have offered a novel and potential treatment method. Compared with conventional radiotherapy, nanotechnology can overcome the blood–brain barrier and improve the sensitivity of glioma to radiotherapy. This paper focuses on the research progress of nano-radiosensitizers in radiotherapy for glioma.
Collapse
Affiliation(s)
- Yandong Xie
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Yuhan Han
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, Suqian First People's Hospital, Suqian, China
| | - Xuefeng Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongwei Ma
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Linfeng Li
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hongmei Liu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
9
|
Ferrari B, Roda E, Priori EC, De Luca F, Facoetti A, Ravera M, Brandalise F, Locatelli CA, Rossi P, Bottone MG. A New Platinum-Based Prodrug Candidate for Chemotherapy and Its Synergistic Effect With Hadrontherapy: Novel Strategy to Treat Glioblastoma. Front Neurosci 2021; 15:589906. [PMID: 33828444 PMCID: PMC8019820 DOI: 10.3389/fnins.2021.589906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 02/08/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) is the most common tumor of the central nervous system. Current therapies, often associated with severe side effects, are inefficacious to contrast the GBM relapsing forms. In trying to overcome these drawbacks, (OC-6-44)-acetatodiamminedichlorido(2-(2-propynyl)octanoato)platinum(IV), also called Pt(IV)Ac-POA, has been recently synthesized. This new prodrug bearing as axial ligand (2-propynyl)octanoic acid (POA), a histone deacetylase inhibitor, has a higher activity due to (i) its high cellular accumulation by virtue of its high lipophilicity and (ii) the inhibition of histone deacetylase, which leads to the increased exposure of nuclear DNA, permitting higher platination and promoting cancer cell death. In the present study, we investigated the effects induced by Pt(IV)Ac-POA and its potential antitumor activity in human U251 glioblastoma cell line using a battery of complementary techniques, i.e., flow cytometry, immunocytochemistry, TEM, and Western blotting analyses. In addition, the synergistic effect of Pt(IV)Ac-POA associated with the innovative oncological hadrontherapy with carbon ions was investigated, with the aim to identify the most efficient anticancer treatment combination. Our in vitro data demonstrated that Pt(IV)Ac-POA is able to induce cell death, through different pathways, at concentrations lower than those tested for other platinum analogs. In particular, an enduring Pt(IV)Ac-POA antitumor effect, persisting in long-term treatment, was demonstrated. Interestingly, this effect was further amplified by the combined exposure to carbon ion radiation. In conclusion, Pt(IV)Ac-POA represents a promising prodrug to be incorporated into the treatment regimen for GBM. Moreover, the synergistic efficacy of the combined protocol using chemotherapeutic Pt(IV)Ac-POA followed by carbon ion radiation may represent a promising approach, which may overcome some typical limitations of conventional therapeutic protocols for GBM treatment.
Collapse
Affiliation(s)
- Beatrice Ferrari
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Elisa Roda
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy.,Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Erica Cecilia Priori
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Fabrizio De Luca
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Angelica Facoetti
- National Center of Oncological Hadrontherapy (Fondazione CNAO), Pavia, Italy
| | - Mauro Ravera
- Department of Sciences and Technological Innovation (DiSIT), University of Piemonte Orientale "A. Avogadro", Alessandria, Italy
| | - Federico Brandalise
- Department of Fundamental Neurosciences (NEUFO), University of Geneva, Geneva, Switzerland
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical & Experimental Toxicology, Pavia Poison Centre, National Toxicology Information Centre, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Paola Rossi
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Maria Grazia Bottone
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| |
Collapse
|
10
|
The HIF1α/JMY pathway promotes glioblastoma stem-like cell invasiveness after irradiation. Sci Rep 2020; 10:18742. [PMID: 33128011 PMCID: PMC7603339 DOI: 10.1038/s41598-020-75300-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 10/13/2020] [Indexed: 01/28/2023] Open
Abstract
Human glioblastoma (GBM) is the most common primary malignant brain tumor. A minor subpopulation of cancer cells, known as glioma stem-like cells (GSCs), are thought to play a major role in tumor relapse due to their stem cell-like properties, their high resistance to conventional treatments and their high invasion capacity. We show that ionizing radiation specifically enhances the motility and invasiveness of human GSCs through the stabilization and nuclear accumulation of the hypoxia-inducible factor 1α (HIF1α), which in turn transcriptionally activates the Junction-mediating and regulatory protein (JMY). Finally, JMY accumulates in the cytoplasm where it stimulates GSC migration via its actin nucleation-promoting activity. Targeting JMY could thus open the way to the development of new therapeutic strategies to improve the efficacy of radiotherapy and prevent glioma recurrence.
Collapse
|
11
|
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
|
12
|
Kong L, Wu J, Gao J, Qiu X, Yang J, Hu J, Hu W, Mao Y, Lu JJ. Particle radiation therapy in the management of malignant glioma: Early experience at the Shanghai Proton and Heavy Ion Center. Cancer 2020; 126:2802-2810. [PMID: 32167589 PMCID: PMC7317504 DOI: 10.1002/cncr.32828] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/20/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022]
Abstract
Background The objective of this study was to evaluate the outcomes of patients with high‐grade glioma who received treatment with particle radiotherapy. Methods Between June 2015 and October 2018, 50 consecutive and nonselected patients with glioblastoma multiforme (n = 34) or anaplastic glioma (n = 16) were treated at the Shanghai Proton and Heavy Ion Center. Twenty‐four patients received proton radiotherapy (at a dose of 60 gray‐equivalents in 30 daily fractions), and 26 patients received proton radiotherapy plus a carbon‐ion radiotherapy (CIRT) boost in various dose‐escalating schemes. All patients received temozolomide because of their age or their O‐6‐methylguanine‐DNA methyltransferase (MGMT) promoter methylation status. Progression‐free survival (PFS) and overall survival (OS) rates, as well as treatment‐induced toxicities, were analyzed. Results At a median follow‐up of 14.3 months (range, 4.8‐39.6 months), the 12‐month and 18‐month OS rates were 87.8% (95% CI, 77.6%‐98.0%) and 72.8% (95% CI, 56.7%‐88.9%), respectively, and the 12‐month and 18‐month PFS rates were 74.2% (95% CI, 60.9%‐87.5%) and 59.8% (95% CI, 43.1%‐76.5%), respectively. Univariate analyses revealed that age (>50 vs ≤50 years), World Health Organization grade (3 vs 4), and Karnofsky performance status (>80 vs ≤80) were significant prognosticators for OS, and IDH mutation and World Health Organization grade were significant for predicting PFS. Furthermore, MGMT promoter methylation, performance status, and age showed a trend toward predicting PFS. No significant predictive factors for PFS or OS were identified in multivariate analyses. Twenty‐nine patients experienced grade 1 treatment‐related acute adverse effects, and 11 developed grade 1 (n = 6) or grade 2 (n = 5) late adverse effect of radiation‐induced brain necrosis. No grade 3, 4, or 5 toxicities were observed. Conclusions Particle radiotherapy produced 18‐month OS and PFS rates of 72.8% and 59.8%, respectively, with acceptable adverse effects in patients with high‐grade glioma. Particle radiotherapy at a dose ≥60 gray‐equivalents appears to be safe and potentially effective. Particle radiotherapy with concurrent temozolomide could potentially produce better outcomes than conventional radiotherapy plus temozolomide. Particle radiotherapy to a dose of ≥60 gray‐equivalents with concurrent temozolomide is safe for patients with high‐grade glioma.
Collapse
Affiliation(s)
- Lin Kong
- Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Fudan University Shanghai Cancer Center, Shanghai, China.,Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China
| | - Jinsong Wu
- Department of Neurosurgery, Fudan University Shanghai Huashan Hospital, Shanghai, China
| | - Jing Gao
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Xianxin Qiu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jing Yang
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Jiyi Hu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Weixu Hu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Fudan University Shanghai Huashan Hospital, Shanghai, China
| | - Jiade J Lu
- Shanghai Engineering Research Center of Proton and Heavy Ion Radiation Therapy, Shanghai, China.,Department of Radiation Oncology, Shanghai Proton and Heavy Ion Center, Shanghai, China
| |
Collapse
|
13
|
Carbon ion radiotherapy in the treatment of gliomas: a review. J Neurooncol 2019; 145:191-199. [DOI: 10.1007/s11060-019-03303-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022]
|
14
|
Shahhoseini E, Feltis BN, Nakayama M, Piva TJ, Pouniotis D, Alghamdi SS, Geso M. Combined Effects of Gold Nanoparticles and Ionizing Radiation on Human Prostate and Lung Cancer Cell Migration. Int J Mol Sci 2019; 20:ijms20184488. [PMID: 31514328 PMCID: PMC6770098 DOI: 10.3390/ijms20184488] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/04/2019] [Accepted: 09/10/2019] [Indexed: 01/15/2023] Open
Abstract
The effect of 15 nm-sized gold nanoparticles (AuNPs) and/or ionizing radiation (IR) on the migration and adhesion of human prostate (DU145) and lung (A549) cancer cell lines was investigated. Cell migration was measured by observing the closing of a gap created by a pipette tip on cell monolayers grown in 6-well plates. The ratio of the gap areas at 0 h and 24 h were used to calculate the relative migration. The relative migration of cells irradiated with 5 Gy was found to be 89% and 86% for DU145 and A549 cells respectively. When the cells were treated with 1 mM AuNPs this fell to ~75% for both cell lines. However, when the cells were treated with both AuNPs and IR an additive effect was seen, as the relative migration rate fell to ~60%. Of interest was that when the cells were exposed to either 2 or 5 Gy IR, their ability to adhere to the surface of a polystyrene culture plate was significantly enhanced, unlike that seen for AuNPs. The delays in gap filling (cell migration) in cells treated with IR and/or AuNPs can be attributed to cellular changes which also may have altered cell motility. In addition, changes in the cytoskeleton of the cancer cells may have also affected adhesiveness and thus the cancer cell's motility response to IR.
Collapse
Affiliation(s)
- Elham Shahhoseini
- Discipline of Medical Radiation, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Bryce N Feltis
- Discipline of Human Bioscience, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Masao Nakayama
- Discipline of Medical Radiation, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Terrence J Piva
- Discipline of Human Bioscience, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Dodie Pouniotis
- Discipline of Laboratory Medicine, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| | - Salem S Alghamdi
- Department of Radiological Sciences, Collage of Applied Medical Science, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia.
| | - Moshi Geso
- Discipline of Medical Radiation, School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Victoria, Australia.
| |
Collapse
|
15
|
Chiblak S, Tang Z, Lemke D, Knoll M, Dokic I, Warta R, Moustafa M, Mier W, Brons S, Rapp C, Muschal S, Seidel P, Bendszus M, Adeberg S, Wiestler OD, Haberkorn U, Debus J, Herold-Mende C, Wick W, Abdollahi A. Carbon irradiation overcomes glioma radioresistance by eradicating stem cells and forming an antiangiogenic and immunopermissive niche. JCI Insight 2019; 4:123837. [PMID: 30674721 DOI: 10.1172/jci.insight.123837] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor radioresistance leading to local therapy failure remains a major obstacle for successful treatment of high-grade glioma. We hypothesized that distinct radiobiological features of particle therapy with carbon ions may circumvent glioma radioresistance. We demonstrate that carbon irradiation (CIR) efficiently eradicates radioresistant patient-derived glioma stem cells (GSCs), leading to growth inhibition and prolonged survival. The impact of CIR at the tumor-stroma interface was further investigated in 2 syngeneic mouse and 2 orthotopic GSC xenograft models. Intriguingly, tumor regressions and long-term local controls were observed at doses greater than or equal to 15-Gy CIR. Fractionated CIR further prolonged survival. The enhanced relative biological effectiveness of CIR in vivo was attributed to its potent antiangiogenic effects and eradication of radioresistant hypoxic tumor cells. Blockade of the HIF1-α/stromal cell-derived factor 1/CXCR4 axis by CIR reduced the recruitment of microglia and myeloid-derived suppressor cells (CD11b+Gr1+). Consequently, CIR abrogated M2-like immune polarization and enhanced the influx of CD8+ cells, generating an immunopermissive niche. We report that radiotherapy with carbon ions could surmount several central glioma resistance mechanisms by eradicating hypoxic and stem cell-like tumor cells, as well as modulating the glioma niche toward an antiangiogenic and less immunosuppressive state. Conclusively, potentially novel rationales for CIR in conquering glioma radioresistance are provided.
Collapse
Affiliation(s)
- Sara Chiblak
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Zili Tang
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Dieter Lemke
- German Cancer Consortium, Heidelberg, Germany.,Department of Neurology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Maximilian Knoll
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Ivana Dokic
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Rolf Warta
- German Cancer Consortium, Heidelberg, Germany.,Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg Medical School, Heidelberg, Germany
| | - Mahmoud Moustafa
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Walter Mier
- German Cancer Consortium, Heidelberg, Germany.,Department of Nuclear Medicine, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Stephan Brons
- Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Carmen Rapp
- German Cancer Consortium, Heidelberg, Germany.,Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg Medical School, Heidelberg, Germany
| | - Stefan Muschal
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Philipp Seidel
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Martin Bendszus
- German Cancer Consortium, Heidelberg, Germany.,Department of Neurology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Sebastian Adeberg
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | | | - Uwe Haberkorn
- German Cancer Consortium, Heidelberg, Germany.,Department of Nuclear Medicine, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Jürgen Debus
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Christel Herold-Mende
- German Cancer Consortium, Heidelberg, Germany.,Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg Medical School, Heidelberg, Germany
| | - Wolfgang Wick
- German Cancer Consortium, Heidelberg, Germany.,Department of Neurology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Amir Abdollahi
- German Cancer Consortium, Heidelberg, Germany.,Division of Molecular & Translational Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology, Heidelberg University Medical School and National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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
|
19
|
Abstract
Carbon ion therapy is a promising evolving modality in radiotherapy to treat tumors that are radioresistant against photon treatments. As carbon ions are more effective in normal and tumor tissue, the relative biological effectiveness (RBE) has to be calculated by bio-mathematical models and has to be considered in the dose prescription. This review (i) introduces the concept of the RBE and its most important determinants, (ii) describes the physical and biological causes of the increased RBE for carbon ions, (iii) summarizes available RBE measurements in vitro and in vivo, and (iv) describes the concepts of the clinically applied RBE models (mixed beam model, local effect model, and microdosimetric-kinetic model), and (v) the way they are introduced into clinical application as well as (vi) their status of experimental and clinical validation, and finally (vii) summarizes the current status of the use of the RBE concept in carbon ion therapy and points out clinically relevant conclusions as well as open questions. The RBE concept has proven to be a valuable concept for dose prescription in carbon ion radiotherapy, however, different centers use different RBE models and therefore care has to be taken when transferring results from one center to another. Experimental studies significantly improve the understanding of the dependencies and limitations of RBE models in clinical application. For the future, further studies investigating quantitatively the differential effects between normal tissues and tumors are needed accompanied by clinical studies on effectiveness and toxicity.
Collapse
Affiliation(s)
- Christian P Karger
- Department of Medical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany. Author to whom any correspondence should be addressed
| | | |
Collapse
|
20
|
Blyth BJ, Cole AJ, MacManus MP, Martin OA. Radiation therapy-induced metastasis: radiobiology and clinical implications. Clin Exp Metastasis 2017; 35:223-236. [PMID: 29159430 DOI: 10.1007/s10585-017-9867-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 11/11/2017] [Indexed: 12/19/2022]
Abstract
Radiation therapy is an effective means of achieving local control in a wide range of primary tumours, with the reduction in the size of the tumour(s) thought to mediate the observed reductions in metastatic spread in clinical trials. However, there is evidence to suggest that the complex changes induced by radiation in the tumour environment can also present metastatic risks that may counteract the long-term efficacy of the treatment. More than 25 years ago, several largely theoretical mechanisms by which radiation exposure might increase metastatic risk were postulated. These include the direct release of tumour cells into the circulation, systemic effects of tumour and normal tissue irradiation and radiation-induced changes in tumour cell phenotype. Here, we review the data that has since emerged to either support or refute these putative mechanisms focusing on how the unique radiobiology underlying modern radiotherapy modalities might alter these risks.
Collapse
Affiliation(s)
- Benjamin J Blyth
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia. .,Cancer Research Division, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia.
| | - Aidan J Cole
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia.,Centre for Cancer Research and Cell Biology, Queen's University Belfast, Lisburn Road, Belfast, BT9 7BL, UK
| | - Michael P MacManus
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Olga A Martin
- Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia.,Cancer Research Division, Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne, 3000, Australia.,The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, 3010, Australia
| |
Collapse
|
21
|
Yamauchi Y, Safi S, Orschiedt L, Gardyan A, Brons S, Rieber J, Nicolay NH, Huber PE, Eichhorn M, Dienemann H, Herth FJF, Weber KJ, Debus J, Hoffmann H, Rieken S. Low-dose photon irradiation induces invasiveness through the SDF-1α/CXCR4 pathway in malignant mesothelioma cells. Oncotarget 2017; 8:68001-68011. [PMID: 28978091 PMCID: PMC5620231 DOI: 10.18632/oncotarget.19134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 06/10/2017] [Indexed: 11/28/2022] Open
Abstract
Background Low-dose photon irradiation has repeatedly been suspected to increase a risk of promoting local recurrence of disease or even systemic dissemination. The purpose of this study was to investigate the motility of malignant pleural mesothelioma (MPM) cell lines after low-doses of photon irradiation and to elucidate the mechanism of the detected phenotype. Methods H28 and H226 MPM cells were examined in clonogenic survival experiments and migration assays with and without various doses of photon and carbon ion irradiation. C-X-C chemokine receptor type 4 (CXCR4), SDF-1α, β1 integrin, α3 integrin, and α5 integrin expressions were analyzed by quantitative FACS analysis, ELISA and western blots. Apoptosis was assessed via Annexin-V-staining. Results The migration of MPM cells was stimulated by both fetal bovine serum and by stromal cell-derived factor 1α (SDF-1α). Low doses of photon irradiation (1 Gy and 2 Gy) suppressed clonogenicity, but promoted migration of both H28 and H226 cells through the SDF-1α/CXCR4 pathway. Hypermigration was inhibited by the administration of CXCR4 antagonist, AMD3100. In contrast, corresponding doses of carbon ion irradiation (0.3 Gy and 1 Gy) suppressed clonogenicity, but did not promote MPM cell migration. Conclusion Our findings suggest that the co-administration of photon irradiation and the CXCR4-antagonist AMD3100 or the use of carbon ions instead of photons may be possible solutions to reduce the risk of locoregional tumor recurrence after radiotherapy for MPM.
Collapse
Affiliation(s)
- Yoshikane Yamauchi
- Department of Thoracic Surgery, Thorax Clinic, Heidelberg University, Heidelberg, Germany
| | - Seyer Safi
- Department of Thoracic Surgery, Thorax Clinic, Heidelberg University, Heidelberg, Germany
| | - Lena Orschiedt
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Adriane Gardyan
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Stephan Brons
- Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Juliane Rieber
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Peter E Huber
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany.,Department of Molecular and Radiation Oncology, German Cancer Research Center (dkfz), Heidelberg, Germany
| | - Martin Eichhorn
- Department of Thoracic Surgery, Thorax Clinic, Heidelberg University, Heidelberg, Germany
| | - Hendrik Dienemann
- Department of Thoracic Surgery, Thorax Clinic, Heidelberg University, Heidelberg, Germany
| | - Felix J F Herth
- Pneumology and Critical Care Medicine, Thorax Clinic, Heidelberg University, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRCH), Heidelberg, Germany, Member of the German Center for Lung Research (DZL)
| | - Klaus-Josef Weber
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| | - Hans Hoffmann
- Department of Thoracic Surgery, Thorax Clinic, Heidelberg University, Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University Hospital of Heidelberg, Heidelberg, Germany.,Heidelberg Ion Treatment Facility (HIT), Heidelberg, Germany.,Heidelberg Institute of Radiation Oncology (HIRO), Heidelberg, Germany
| |
Collapse
|
22
|
Winter M, Dokic I, Schlegel J, Warnken U, Debus J, Abdollahi A, Schnölzer M. Deciphering the Acute Cellular Phosphoproteome Response to Irradiation with X-rays, Protons and Carbon Ions. Mol Cell Proteomics 2017; 16:855-872. [PMID: 28302921 DOI: 10.1074/mcp.m116.066597] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/15/2017] [Indexed: 12/15/2022] Open
Abstract
Radiotherapy is a cornerstone of cancer therapy. The recently established particle therapy with raster-scanning protons and carbon ions landmarks a new era in the field of high-precision cancer medicine. However, molecular mechanisms governing radiation induced intracellular signaling remain elusive. Here, we present the first comprehensive proteomic and phosphoproteomic study applying stable isotope labeling by amino acids in cell culture (SILAC) in combination with high-resolution mass spectrometry to decipher cellular response to irradiation with X-rays, protons and carbon ions. At protein expression level limited alterations were observed 2 h post irradiation of human lung adenocarcinoma cells. In contrast, 181 phosphorylation sites were found to be differentially regulated out of which 151 sites were not hitherto attributed to radiation response as revealed by crosscheck with the PhosphoSitePlus database.Radiation-induced phosphorylation of the p(S/T)Q motif was the prevailing regulation pattern affecting proteins involved in DNA damage response signaling. Because radiation doses were selected to produce same level of cell kill and DNA double-strand breakage for each radiation quality, DNA damage responsive phosphorylation sites were regulated to same extent. However, differential phosphorylation between radiation qualities was observed for 55 phosphorylation sites indicating the existence of distinct signaling circuitries induced by X-ray versus particle (proton/carbon) irradiation beyond the canonical DNA damage response. This unexpected finding was confirmed in targeted spike-in experiments using synthetic isotope labeled phosphopeptides. Herewith, we successfully validated uniform DNA damage response signaling coexisting with altered signaling involved in apoptosis and metabolic processes induced by X-ray and particle based treatments.In summary, the comprehensive insight into the radiation-induced phosphoproteome landscape is instructive for the design of functional studies aiming to decipher cellular signaling processes in response to radiotherapy, space radiation or ionizing radiation per se Further, our data will have a significant impact on the ongoing debate about patient treatment modalities.
Collapse
Affiliation(s)
- Martin Winter
- From the ‡Functional Proteome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany.,§Translational Radiation Oncology, National Center for Tumor diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany.,¶German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Ivana Dokic
- §Translational Radiation Oncology, National Center for Tumor diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany.,¶German Cancer Consortium (DKTK), Heidelberg, Germany.,‖Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 450, D-69120 Heidelberg, Germany.,**Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Julian Schlegel
- §Translational Radiation Oncology, National Center for Tumor diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany.,¶German Cancer Consortium (DKTK), Heidelberg, Germany.,‖Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 450, D-69120 Heidelberg, Germany.,**Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Uwe Warnken
- From the ‡Functional Proteome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany
| | - Jürgen Debus
- §Translational Radiation Oncology, National Center for Tumor diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany.,¶German Cancer Consortium (DKTK), Heidelberg, Germany.,‖Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 450, D-69120 Heidelberg, Germany.,**Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Amir Abdollahi
- §Translational Radiation Oncology, National Center for Tumor diseases (NCT), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 460, D-69120 Heidelberg, Germany.,¶German Cancer Consortium (DKTK), Heidelberg, Germany.,‖Heidelberg Ion Beam Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 450, D-69120 Heidelberg, Germany.,**Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), Heidelberg, Germany
| | - Martina Schnölzer
- From the ‡Functional Proteome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, D-69120 Heidelberg, Germany;
| |
Collapse
|
23
|
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
|
24
|
Fujita M, Yamada S, Imai T. Irradiation induces diverse changes in invasive potential in cancer cell lines. Semin Cancer Biol 2015; 35:45-52. [DOI: 10.1016/j.semcancer.2015.09.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 12/14/2022]
|
25
|
Laprie A, Hu Y, Alapetite C, Carrie C, Habrand JL, Bolle S, Bondiau PY, Ducassou A, Huchet A, Bertozzi AI, Perel Y, Moyal É, Balosso J. Paediatric brain tumours: A review of radiotherapy, state of the art and challenges for the future regarding protontherapy and carbontherapy. Cancer Radiother 2015; 19:775-89. [PMID: 26548600 DOI: 10.1016/j.canrad.2015.05.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 05/18/2015] [Accepted: 05/21/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND PURPOSE Brain tumours are the most frequent solid tumours in children and the most frequent radiotherapy indications in paediatrics, with frequent late effects: cognitive, osseous, visual, auditory and hormonal. A better protection of healthy tissues by improved beam ballistics, with particle therapy, is expected to decrease significantly late effects without decreasing local control and survival. This article reviews the scientific literature to advocate indications of protontherapy and carbon ion therapy for childhood central nervous system cancer, and estimate the expected therapeutic benefits. MATERIALS AND METHODS A systematic review was performed on paediatric brain tumour treatments using Medline (from 1966 to March of 2014). To be included, clinical trials had to meet the following criteria: age of patients 18 years or younger, treated with radiation, and report of survival. Studies were also selected according to the evidence level. A secondary search of cited references found other studies about cognitive functions, quality of life, the comparison of photon and proton dosimetry showing potential dose escalation and/or sparing of organs at risk with protontherapy; and studies on dosimetric and technical issues related to protontherapy. RESULTS A total of 7051 primary references published were retrieved, among which 40 clinical studies and 60 papers about quality of life, dose distribution and dosimetry were analysed, as well as the ongoing clinical trials. These papers have been summarized and reported in a specific document made available to the participants of a final 1-day workshop. Tumours of the meningeal envelop and bony cranial structures were excluded from the analysis. Protontherapy allows outstanding ballistics to target the tumour area, while substantially decreasing radiation dose to the normal tissues. There are many indications of protontherapy for paediatric brain tumours in curative intent, either for localized treatment of ependymomas, germ-cell tumours, craniopharyngiomas, low-grade gliomas; or panventricular irradiation of pure non-secreting germinoma; or craniospinal irradiation of medulloblastomas and metastatic pure germinomas. Carbon ion therapy is just emerging and may be studied for highly aggressive and radioresistant tumours, as an initial treatment for diffuse brainstem gliomas, and for relapse of high-grade gliomas. CONCLUSION Both protontherapy and carbon ion therapy are promising for paediatric brain tumours. The benefit of decreasing late effects without altering survival has been described for most paediatric brain tumours with protontherapy and is currently assessed in ongoing clinical trials with up-to-date proton devices. Unfortunately, in 2015, only a minority of paediatric patients in France can receive protontherapy due to the lack of equipment.
Collapse
Affiliation(s)
- A Laprie
- Université Paul-Sabatier, Toulouse, France; Institut Claudius-Regaud, institut universitaire du cancer de Toulouse (IUCT)-Oncopole, radiation oncology, 1, avenue Irene-Joliot-Curie, 31059 Toulouse, France; Périclès-France-Hadron, Toulouse, France.
| | - Y Hu
- GCS-Étoile-France-Hadron, Lyon, France
| | - C Alapetite
- Institut Curie Paris Orsay (ICPO)-France-Hadron, Orsay, France
| | - C Carrie
- GCS-Étoile-France-Hadron, Lyon, France; Centre Léon-Bérard, Lyon, France
| | - J-L Habrand
- Institut Curie Paris Orsay (ICPO)-France-Hadron, Orsay, France; Université Paris Sud, Orsay, France; Archade-France-Hadron, Caen, France; Centre François-Baclesse, Caen, France; Gustave-Roussy, Villejuif, France
| | - S Bolle
- Institut Curie Paris Orsay (ICPO)-France-Hadron, Orsay, France; Impact-France-Hadron, Nice, France
| | - P-Y Bondiau
- Centre Antoine-Lacassagne, Nice, France; CHU de Bordeaux, Bordeaux, France
| | - A Ducassou
- Institut Claudius-Regaud, institut universitaire du cancer de Toulouse (IUCT)-Oncopole, radiation oncology, 1, avenue Irene-Joliot-Curie, 31059 Toulouse, France; Périclès-France-Hadron, Toulouse, France
| | - A Huchet
- Hôpital des Enfants, Toulouse, France
| | - A-I Bertozzi
- Périclès-France-Hadron, Toulouse, France; Université Grenoble Alpes, Grenoble, France
| | - Y Perel
- Université Grenoble Alpes, Grenoble, France
| | - É Moyal
- Université Paul-Sabatier, Toulouse, France; Institut Claudius-Regaud, institut universitaire du cancer de Toulouse (IUCT)-Oncopole, radiation oncology, 1, avenue Irene-Joliot-Curie, 31059 Toulouse, France; Périclès-France-Hadron, Toulouse, France
| | - J Balosso
- GCS-Étoile-France-Hadron, Lyon, France; CHU de Grenoble, Grenoble, France
| |
Collapse
|
26
|
Rieken S, Rieber J, Brons S, Habermehl D, Rief H, Orschiedt L, Lindel K, Weber KJ, Debus J, Combs SE. Radiation-induced motility alterations in medulloblastoma cells. JOURNAL OF RADIATION RESEARCH 2015; 56:430-436. [PMID: 25736470 PMCID: PMC4426914 DOI: 10.1093/jrr/rru120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/09/2014] [Accepted: 11/21/2014] [Indexed: 05/28/2023]
Abstract
Photon irradiation has been repeatedly suspected of increasing tumor cell motility and promoting locoregional recurrence of disease. This study was set up to analyse possible mechanisms underlying the potentially radiation-altered motility in medulloblastoma cells. Medulloblastoma cell lines D425 and Med8A were analyzed in migration and adhesion experiments with and without photon and carbon ion irradiation. Expression of integrins was determined by quantitative FACS analysis. Matrix metalloproteinase concentrations within cell culture supernatants were investigated by enzyme-linked immunosorbent assay (ELISA). Statistical analysis was performed using Student's t-test. Both photon and carbon ion irradiation significantly reduced chemotactic medulloblastoma cell transmigration through 8-μm pore size membranes, while simultaneously increasing adherence to fibronectin- and collagen I- and IV-coated surfaces. Correspondingly, both photon and carbon ion irradiation downregulate soluble MMP9 concentrations, while upregulating cell surface expression of proadhesive extracellular matrix protein-binding integrin α5. The observed phenotype of radiation-altered motility is more pronounced following carbon ion than photon irradiation. Both photon and (even more so) carbon ion irradiation are effective in inhibiting medulloblastoma cell migration through downregulation of matrix metalloproteinase 9 and upregulation of proadhesive cell surface integrin α5, which lead to increased cell adherence to extracellular matrix proteins.
Collapse
Affiliation(s)
- Stefan Rieken
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Juliane Rieber
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Stephan Brons
- Heidelberg Ion Treatment Facility (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
| | - Daniel Habermehl
- Klinikum rechts der Isar, Technische Universität München, Klinik für Radioonkologie und Strahlentherapie, Ismaninger Straße 22, 81675 München, Germany
| | - Harald Rief
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Lena Orschiedt
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Katja Lindel
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Klaus J Weber
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Jürgen Debus
- University Hospital of Heidelberg, Department of Radiation Oncology, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Stephanie E Combs
- Klinikum rechts der Isar, Technische Universität München, Klinik für Radioonkologie und Strahlentherapie, Ismaninger Straße 22, 81675 München, Germany
| |
Collapse
|
27
|
Abstract
Glioblastoma multiform is the most common and aggressive brain tumor with a worse prognostic. Ionizing radiation is a cornerstone in the treatment of glioblastome with chemo-radiation association being the actual standard. As a paradoxal effect, it has been suggested that radiotherapy could have a deleterious effect on local recurrence of cancer. In vivo studies have studied the effect of radiotherapy on biological modification and pathogenous effect of cancer cells. It seems that ionizing radiations with photon could activate oncogenic pathways in glioblastoma cell lines. We realized a review of the literature of photon-enhanced effect on invasion and migration of glioblastoma cells by radiotherapy.
Collapse
|
28
|
Simon F, Dittmar JO, Brons S, Orschiedt L, Urbschat S, Weber KJ, Debus J, Combs SE, Rieken S. Integrin-based meningioma cell migration is promoted by photon but not by carbon-ion irradiation. Strahlenther Onkol 2014; 191:347-55. [PMID: 25445155 DOI: 10.1007/s00066-014-0778-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/29/2014] [Indexed: 12/11/2022]
Abstract
PURPOSE Sublethal doses of photon irradiation (IR) are suspected to increase tumor cell migration and support locoregional recurrence of disease, which has already been shown in other cell lines. This manuscript describes the effect of photon and carbon-ion IR on WHO class I meningioma cell migration and provides an approach to the underlying cellular mechanisms. MATERIALS AND METHODS Meningioma cells were gained operatively at the university hospital in Homburg/Saar, Germany. For migration, membranes (8-µm pore sizes) were coated with collagen I, with collagen IV, and with fibronectin. Cells were analyzed in migration experiments with or without serum stimulation, with or without photon and carbon IR 24 h prior to experiments, and with or without integrin antibodies. Fluorescence-activated cell sorting (FACS) analyses of the integrins ανβ1, ανβ3, and ανβ5 were performed without IR and 6, 12 and 24 h after IR. Enzyme-linked immunosorbent assay (ELISA) analyses of matrix metalloproteinases (MMP)-2 and MMP-9 were realized with and without IR after cells were cultured on collagen I, collagen IV, or fibronectin for 24 h. Cells and supernatants for FACS and ELISA were stored at - 18 °C. The significance level was set at 5 % using both Student's t test and two-way ANOVA. RESULTS Migration of meningioma cells was serum-inducible (p < 0.001). It could be increased by photon IR (p < 0.02). The integrins ανβ1 and ανβ5 showed a 21 and 11 % higher expression after serum stimulation (not significant), respectively, and ανβ1 expression was raised by 14 % (p = 0.0057) after photon IR. Antibody blockage of the integrins ανβ1 and ανβ5 inhibited serum- and photon-induced migration. Expression of MMP-2 and MMP-9 remained unchanged after both IR and fetal bovine serum (FBS). Carbon-ion IR left both integrin expression and meningioma cell migration unaffected. CONCLUSION Photon but not carbon-ion IR promotes serum-based meningioma cell migration. Fibronectin receptor integrin ανβ1 signaling can be identified as an important mechanism for serum- and photon-induced migration of WHO class I meningioma cells.
Collapse
Affiliation(s)
- Florian Simon
- Department of Radiation Oncology, University Hospital of Heidelberg, Im Neuenheimer Feld 400, 69120, Heidelberg, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Perona M, Dagrosa MA, Pagotto R, Casal M, Pignataro O, Pisarev MA, Juvenal GJ. Protective effect of an antithyroid compound against γ-radiation-induced damage in human colon cancer cells. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2014; 53:611-619. [PMID: 24811726 DOI: 10.1007/s00411-014-0542-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Accepted: 04/13/2014] [Indexed: 06/03/2023]
Abstract
We have previously reported the radioprotective effect of propylthiouracil (PTU) on thyroid cells. The aim of the present study was to analyze whether tumor cells and normal cells demonstrate the same response to PTU. Human colon carcinoma cells were irradiated with γ-irradiation with or without PTU. We evaluated the clonogenic survival, intracellular reactive oxygen species levels, catalase, superoxide dismutase and glutathione peroxidase activities, and apoptosis by nuclear cell morphology and caspase-3 activity assays. Cyclic AMP (cAMP) levels were measured by radioimmunoassay. PTU treatment increased surviving cell fraction at 2 Gy (SF2) from 56.9 ± 3.6 in controls to 75.0 ± 3.5 (p < 0.05) and diminished radiation-induced apoptosis. In addition, we observed that the level of antioxidant enzymes' activity was increased in cells treated with PTU. Moreover, pretreatment with PTU increased intracellular levels of cAMP. Forskolin (p < 0.01) and dibutyryl cAMP (p < 0.05) mimicked the effect of PTU on SF2. Co-treatment with H89, an inhibitor of protein kinase A, abolished the radioprotective effect of PTU. PTU reduces the toxicity of ionizing radiation by increasing cAMP levels and also possibly through a reduction in apoptosis levels and in radiation-induced oxidative stress damage. We therefore conclude that PTU protects both normal and cancer cells during exposure to radiation in conditions mimicking the radiotherapy.
Collapse
Affiliation(s)
- Marina Perona
- Department of Radiobiology (CAC), National Atomic Energy Commission (CNEA), San Martín, Buenos Aires, Argentina
| | | | | | | | | | | | | |
Collapse
|
30
|
Moncharmont C, Levy A, Guy JB, Falk AT, Guilbert M, Trone JC, Alphonse G, Gilormini M, Ardail D, Toillon RA, Rodriguez-Lafrasse C, Magné N. Radiation-enhanced cell migration/invasion process: a review. Crit Rev Oncol Hematol 2014; 92:133-42. [PMID: 24908570 DOI: 10.1016/j.critrevonc.2014.05.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 04/25/2014] [Accepted: 05/09/2014] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy is a keystone treatment in cancer. Photon radiation has proved its benefits in overall survival in many clinical studies. However, some patients present local recurrences or metastases when cancer cells survive to treatment. Metastasis is a process which includes adhesion of the cell to the extracellular matrix, degradation of the matrix by proteases, cell motility, intravasation in blood or lymphatic vessels, extravasation in distant parenchyma and development of cell colonies. Several studies demonstrated that ionizing radiation might promote migration and invasion of tumor cells by intricate implications in the micro-environment, cell-cell junctions, extracellular matrix junctions, proteases secretion, and induction of epithelial-mesenchymal transition. This review reports various cellular pathways involved in the photon-enhanced cell invasion process for which potential therapeutic target may be employed for enhancing antitumor effectiveness. Understanding these mechanisms could lead to therapeutic strategies to counter the highly invasive cell lines via specific inhibitors or carbon-ion therapy.
Collapse
Affiliation(s)
- Coralie Moncharmont
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France; Department of Radiotherapy, Institut de Cancérologie de la Lucien Neuwirth, St Priest en Jarez, France
| | - Antonin Levy
- Department of Radiotherapy, GustaveRoussy, Villejuif, France
| | - Jean-Baptiste Guy
- Department of Radiotherapy, Institut de Cancérologie de la Lucien Neuwirth, St Priest en Jarez, France
| | - Alexander T Falk
- Department of Radiotherapy, Centre Antoine Lacassagne, Nice, France
| | - Matthieu Guilbert
- INSERM U908, Growth Factor Signalling in Breast Cancer, Functional Proteomics, University Lille 1, IFR-147, 59000 Villeneuve d'Ascq, France
| | - Jane-Chloé Trone
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France
| | - Gersende Alphonse
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France
| | - Marion Gilormini
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France
| | - Dominique Ardail
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France
| | - Robert-Alain Toillon
- INSERM U908, Growth Factor Signalling in Breast Cancer, Functional Proteomics, University Lille 1, IFR-147, 59000 Villeneuve d'Ascq, France
| | - Claire Rodriguez-Lafrasse
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France
| | - Nicolas Magné
- Laboratoire de Radiobiologie Cellulaire et Moléculaire, EMR3738, Faculté de Médecine Lyon Sud, 69921 Oullins, France; Department of Radiotherapy, Institut de Cancérologie de la Lucien Neuwirth, St Priest en Jarez, France.
| |
Collapse
|
31
|
Karger CP, Scholz M, Huber PE, Debus J, Peschke P. Photon and carbon ion irradiation of a rat prostate carcinoma: does a higher fraction number increase the metastatic rate? Radiat Res 2014; 181:623-8. [PMID: 24844648 DOI: 10.1667/rr13611.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
In a recent published study, we investigated the response of an experimental prostate carcinoma (R3327-AT1) after irradiation with 1, 2 or 6 fractions of carbon ions or photons, respectively. The original intention of this study was to measure the dose-dependent local control probability as well as the related relative biological effectiveness of carbon ions. However, we now report an increased metastatic rate when the number of fractions was increased from 2 to 6. In a total of 246 animals, the actuarial metastatic rates for 1, 2 and 6 fractions were 5.1 ± 3.5%, 5.7 ± 4.0% and 15.3 ± 7.1% for photons and 9.8 ± 7.5%, 4.0 ± 3.9% and 20.3 ± 6.5% for carbon ions, respectively. The increase was significant only for carbon ions (6 vs. 2 fractions,P = 0.03). Although the original experiment was not designed to investigate metastatic rates, this observation may be of general interest to researchers studying radiation-modulated metastatic activity.
Collapse
Affiliation(s)
- Christian P Karger
- a Department of Medical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | | | | |
Collapse
|
32
|
Debus J, Abdollahi A. For the next trick: new discoveries in radiobiology applied to glioblastoma. Am Soc Clin Oncol Educ Book 2014:e95-e99. [PMID: 24857153 DOI: 10.14694/edbook_am.2014.34.e95] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor. Radiotherapy post surgical resection remained the mainstay of the management of GBM for decades until the addition of temozolomide was shown to prolong the median overall survival (OS) by 2.5 months to 14.6 months in 2005. Infiltrative growth to surrounding normal brain tissue and cooption of vascular niches, peripheral microvasuclar hyperplasia, and central hypoxic regions with pseudopalisading necrosis are characteristics of GBM and are causally linked to their exceptional radio- and chemo-resistant phenotype. An intratumoral hierarchy is postulated consisting of tumor stem cells in the apex with high DNA-repair proficiency resisting radiotherapy. It is conceivable that the stem cell property is more dynamic than originally anticipated. Niche effects such as exposure to hypoxia and intercellular communication in proximities to endothelial or bone marrow-derived cells (BMDC), for example, may activate such "stem cell" programs. GBM are exceptionally stroma-rich tumors and may consist of more than 70% stroma components, such as microglia and BMDC. It becomes increasingly apparent that treatment of GBM needs to integrate therapies targeting all above-mentioned distinct pathophysiological features. Accordingly, recent approaches in GBM therapy include inhibition of invasion (e.g., integrin, EGFR, CD95, and mTOR inhibition), antiangiogenesis and stroma modulators (TGFbeta, VEGF, angiopoetin, cMET inhibitors) and activation of immune response (vaccination and blockage of negative co-stimulatory signals). In addition, high LET-radiotherapy, for example with carbon ions, is postulated to ablate tumor stem cell and hypoxic cells more efficiently as compared with conventional low-LET photon irradiation. We discuss current key concepts, their limitations, and potentials to improve the outcome in this rapidly progressive and devastating disease.
Collapse
Affiliation(s)
- Juergen Debus
- From the German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School; Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Amir Abdollahi
- From the German Cancer Consortium (DKTK), Heidelberg, Germany; Department of Radiation Oncology, Heidelberg Ion Therapy Center (HIT), Heidelberg Institute of Radiation Oncology (HIRO), University of Heidelberg Medical School; Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
33
|
Combs SE, Debus J. Treatment with heavy charged particles: systematic review of clinical data and current clinical (comparative) trials. Acta Oncol 2013; 52:1272-86. [PMID: 23964656 DOI: 10.3109/0284186x.2013.818254] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BACKGROUND To analyze relevant data on carbon ion radiotherapy for different tumor indications and to review current clinical trials. MATERIAL AND METHODS All published data on carbon ion radiotherapy were searched for with specific criteria in PUBMED. The terms for search were 'carbon ion and (radiotherapy OR radiation therapy) and (nirs OR chiba OR japan OR itep OR st. petersburg OR PSI OR dubna OR uppsala OR clatterbridge OR loma linda OR nice OR orsay OR itemba OR mpri OR himac OR triumf OR GSI OR HMI OR NCC OR ibmc OR pmrc OR MGH OR infn-lns OR shizuoka OR werc OR zibo OR md anderson OR fpti OR ncc ilsan OR boston OR heidelberg OR tsukuba) NOT in vitro NOT cell culture NOT review[Publication Type] Filters: Humans, English'. The search delivered 273 hits, of which only articles in English including 20 or more patients were included. Case reports were not considered. We subdivided into disease- and site-specific groups. RESULTS AND CONCLUSION To date, several studies have been performed, however, no randomized trials have been conducted. Therefore, carbon ion radiotherapy must be considered an experimental treatment, and randomized trials comparing modern photon as well as proton treatments are necessary.
Collapse
Affiliation(s)
- Stephanie E Combs
- University Hospital of Heidelberg, Department of Radiation Oncology , Heidelberg , Germany
| | | |
Collapse
|
34
|
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
|
35
|
El Shafie RA, Habermehl D, Rieken S, Mairani A, Orschiedt L, Brons S, Haberer T, Weber KJ, Debus J, Combs SE. In vitro evaluation of photon and raster-scanned carbon ion radiotherapy in combination with gemcitabine in pancreatic cancer cell lines. JOURNAL OF RADIATION RESEARCH 2013; 54 Suppl 1:i113-i119. [PMID: 23824114 PMCID: PMC3700516 DOI: 10.1093/jrr/rrt052] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 03/26/2013] [Accepted: 04/01/2013] [Indexed: 05/28/2023]
Abstract
BACKGROUND Pancreatic cancer is the fourth leading cause of cancer deaths, being responsible for 6% of all cancer-related deaths. Conventional radiotherapy with or without additional chemotherapy has been applied in the past in the context of neoadjuvant or adjuvant therapy concepts with only modest results, however new radiation modalities, such as particle therapy with promising physical and biological characteristics, present an alternative treatment option for patients with pancreatic cancer. Up until now the raster scanning technique employed at our institution for the application of carbon ions has been unique, and no radiobiological data using pancreatic cancer cells has been available yet. The aim of this study was to evaluate cytotoxic effects that can be achieved by treating pancreatic cancer cell lines with combinations of X-rays and gemcitabine, or alternatively with carbon ion irradiation and gemcitabine, respectively. MATERIALS AND METHODS Human pancreatic cancer cell lines AsPC-1, BxPC-3 and Panc-1 were irradiated with photons and carbon ions at various doses and treated with gemcitabine. Photon irradiation was applied with a biological cabin X-ray irradiator, and carbon ion irradiation was applied with an extended Bragg peak (linear energy transfer (LET) 103 keV/μm) using the raster scanning technique at the Heidelberg Ion Therapy Center (HIT). Responsiveness of pancreatic cancer cells to the treatment was measured by clonogenic survival. Clonogenic survival curves were then compared to predicted curves that were calculated employing the local effect model (LEM). RESULTS Cell survival curves were calculated from the surviving fractions of each combination experiment and compared to a drug control that was only irradiated with X-rays or carbon ions, without application of gemcitabine. In terms of cytotoxicity, additive effects were achieved for the cell lines Panc-1 and BxPC-3, and a slight radiosensitizing effect was observed for AsPC-1. Relative biological effectiveness (RBE) of carbon ion irradiation ranged from 1.5-4.5 depending on survival level and dose. Sensitizer enhancement ratio (SER) values calculated at 10% cell survival ranged from 1.24-1.66, depending on cell line, gemcitabine dose and irradiation modality. Experimentally ascertained survival curves matched those predicted by LEM-calculation. CONCLUSION Our experiments have shown a combined treatment of irradiation and chemotherapy with gemcitabine to be a good means of achieving additive cytotoxic effects on pancreatic cancer cell lines. The data generated in this study will serve as radiobiological basis for further preclinical and clinical studies.
Collapse
Affiliation(s)
- Rami A. El Shafie
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Daniel Habermehl
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Stefan Rieken
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Andrea Mairani
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
| | - Lena Orschiedt
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Stefan Brons
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Thomas Haberer
- Heidelberg Ion Therapy Center (HIT), Im Neuenheimer Feld 450, 69120 Heidelberg, Germany
| | - Klaus-Josef Weber
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Jürgen Debus
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, University of Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| |
Collapse
|
36
|
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]
|
37
|
Three-dimensional Invasion of Human Glioblastoma Cells Remains Unchanged by X-ray and Carbon Ion Irradiation In Vitro. Int J Radiat Oncol Biol Phys 2012; 84:e515-23. [DOI: 10.1016/j.ijrobp.2012.06.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 04/27/2012] [Accepted: 06/06/2012] [Indexed: 01/29/2023]
|
38
|
Blakely EA. Lauriston S. Taylor Lecture on radiation protection and measurements: what makes particle radiation so effective? HEALTH PHYSICS 2012; 103:508-28. [PMID: 23032880 PMCID: PMC3507469 DOI: 10.1097/hp.0b013e31826a5b85] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The scientific basis for the physical and biological effectiveness of particle radiations has emerged from many decades of meticulous basic research. A diverse array of biologically relevant consequences at the molecular, cellular, tissue, and organism level have been reported, but what are the key processes and mechanisms that make particle radiation so effective, and what competing processes define dose dependences? Recent studies have shown that individual genotypes control radiation-regulated genes and pathways in response to radiations of varying ionization density. The fact that densely ionizing radiations can affect different gene families than sparsely ionizing radiations, and that the effects are dose- and time-dependent, has opened up new areas of future research. The complex microenvironment of the stroma and the significant contributions of the immune response have added to our understanding of tissue-specific differences across the linear energy transfer (LET) spectrum. The importance of targeted versus nontargeted effects remains a thorny but elusive and important contributor to chronic low dose radiation effects of variable LET that still needs further research. The induction of cancer is also LET-dependent, suggesting different mechanisms of action across the gradient of ionization density. The focus of this 35th Lauriston S. Taylor Lecture is to chronicle the step-by-step acquisition of experimental clues that have refined our understanding of what makes particle radiation so effective, with emphasis on the example of radiation effects on the crystalline lens of the human eye.
Collapse
Affiliation(s)
- Eleanor A Blakely
- Lawrence Berkeley National Laboratory, One Cyclotron Road, MS 977, Berkeley, CA 94720, USA.
| |
Collapse
|
39
|
Ferrandon S, Saultier P, Carras J, Battiston-Montagne P, Alphonse G, Beuve M, Malleval C, Honnorat J, Slatter T, Hung N, Royds J, Rodriguez-Lafrasse C, Poncet D. Telomere profiling: toward glioblastoma personalized medicine. Mol Neurobiol 2012; 47:64-76. [PMID: 23065374 DOI: 10.1007/s12035-012-8363-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 10/01/2012] [Indexed: 02/03/2023]
Abstract
Despite a standard of care combining surgery, radiotherapy (RT), and temozolomide chemotherapy, the average overall survival (OS) of glioblastoma patients is only 15 months, and even far lower when the patient cannot benefit from this combination. Therefore, there is a strong need for new treatments, such as new irradiation techniques. Against this background, carbon ion hadrontherapy, a new kind of irradiation, leads to a greater biological response of the tumor, while minimizing adverse effects on healthy tissues in comparison with RT. As carbon ion hadrontherapy is restricted to RT-resistant patients, photon irradiation resistance biomarkers are needed. Long telomeres and high telomerase activity have been widely associated with photon radioresistance in other cancers. Moreover, telomere protection, telomere function, and telomere length (TL) also depend on the shelterin protein complex (TRF1, TRF2, TPP1, POT1, TIN2, and hRAP1). We thus decided to evaluate an enlarged telomeric status (TL, telomerase catalytic subunit, and the shelterin component expression level) as a potential radioresistance biomarker in vitro using cellular models and ex vivo using patient tumor biopsies. In addition, nothing was known about the role of telomeres in carbon ion response. We thus evaluated telomeric status after both types of irradiation. We report here a significant correlation between TL and the basal POT1 expression level and photon radioresistance, in vitro, and a significant increase in the OS of patients with long telomeres or a high POT1 level, in vivo. POT1 expression was predictive of patient response irrespective of the TL. Strikingly, these correlations were lost, in vitro, when considering carbon irradiation. We thus propose (1) a model of the implications of telomeric damage in the cell response to both types of irradiation and (2) assessment of the POT1 expression level and TL using patient tumor biopsies to identify radioresistant patients who could benefit from carbon hadrontherapy.
Collapse
Affiliation(s)
- Sylvain Ferrandon
- EMR3738, Cellular and Molecular Radiobiology Laboratory, Medicine Faculty, Lyon 1 University, 69921, Oullins Cedex 12, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Azria D, Ardiet JM, Chauvet B, Denis F, Eschwège F, Hennequin C, Lartigau É, Rocher F, Mahé MA, Maingon P, Mazeron JJ, Metayer Y, Peiffert D, Thureau S, Mornex F. Implications récentes des équipes françaises en oncologie radiothérapie et radiobiologie clinique. Cancer Radiother 2012; 16:386-91. [DOI: 10.1016/j.canrad.2012.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 05/18/2012] [Indexed: 12/30/2022]
|