1
|
Marinescu IM, Rogg M, Spohn S, von Büren M, Kamps M, Jilg CA, Fountzila E, Papadopoulou K, Ceci L, Bettermann A, Ruf J, Benndorf M, Adebahr S, Zips D, Grosu AL, Schell C, Zamboglou C. Ex vivo γH2AX assay for tumor radiosensitivity in primary prostate cancer patients and correlation with clinical parameters. Radiat Oncol 2022; 17:163. [PMID: 36199143 PMCID: PMC9533509 DOI: 10.1186/s13014-022-02131-1] [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: 08/22/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Backround Accurate surrogate parameters for radio resistance are warranted for individualized radiotherapy (RT) concepts in prostate cancer (PCa). The purpose of this study was to assess intertumoral heterogeneity in terms of radio resistance using an ex-vivo γH2AX assay after irradiation of prostate biopsy cores and to investigate its correlation with clinical features of respective patients as well as imaging and genomic features of tumor areas.
Methods Twenty one patients with histologically-proven PCa and pre-therapeutic multiparametric resonance imaging and prostate-specific membrane antigen positron emission tomography were included in the study. Biopsy cores were collected from 26 PCa foci. Residual γH2AX foci were counted 24 h after ex-vivo irradiation (with 0 and 4 Gy) of biopsy specimen and served as a surrogate for radio resistance. Clinical, genomic (next generation sequencing) and imaging features were collected and their association with the radio resistance was studied. Results In total 18 PCa lesions from 16 patients were included in the final analysis. The median γH2AX foci value per PCa lesion was 3.12. According to this, the patients were divided into two groups (radio sensitive vs. radio resistant) with significant differences in foci number (p < 0.0001). The patients in the radio sensitive group had significantly higher prostate specific antigen serum concentration (p = 0.015), tumor areas in the radio sensitive group had higher SUV (standardized uptake values in PSMA PET)-max and -mean values (p = 0.0037, p = 0.028) and lower ADC (apparent diffusion coefficient-mean values, p = 0.049). All later parameters had significant (p < 0.05) correlations in Pearson’s test. One patient in the radio sensitive group displayed a previously not reported loss of function frameshift mutation in the NBN gene (c.654_658delAAAAC) that introduces a premature termination codon and results in a truncated protein. Conclusion In this pilot study, significant differences in intertumoral radio resistance were observed and clinical as well as imaging parameters may be applied for their prediction. After further prospective validation in larger patient cohorts these finding may lead to individual RT dose prescription for PCa patients in the future.
Collapse
Affiliation(s)
- Ioana M Marinescu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany.
| | - Manuel Rogg
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Simon Spohn
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Moritz von Büren
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Marius Kamps
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Cordula A Jilg
- Department of Urology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Elena Fountzila
- Second Department of Medical Oncology, Euromedica General Clinic of Thessaloniki, Thessaloniki, Greece.,Greece and European University Cyprus, Engomi, Cyprus
| | - Kyriaki Papadopoulou
- Laboratory of Molecular Oncology, Hellenic Foundation for Cancer Research, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lara Ceci
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Alisa Bettermann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Matthias Benndorf
- Department of Radiology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Sonja Adebahr
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Daniel Zips
- Medical Faculty and University Hospital, Radiation Oncology, Eberhard Karls University Tübingen, Tübingen, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany
| | - Christoph Schell
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site, Freiburg, Germany.,Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Tumorbank Comprehensive Cancer Center Freiburg, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, University of Freiburg, Freiburg, Germany.,German Oncology Center, European University Cyprus, Limassol, Cyprus
| |
Collapse
|
2
|
CD44, γ-H2AX, and p-ATM Expressions in Short-Term Ex Vivo Culture of Tumour Slices Predict the Treatment Response in Patients with Oral Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23020877. [PMID: 35055060 PMCID: PMC8775909 DOI: 10.3390/ijms23020877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
Squamous cell carcinoma is the most common type of head and neck cancer (HNSCC) with a disease-free survival at 3 years that does not exceed 30%. Biomarkers able to predict clinical outcomes are clearly needed. The purpose of this study was to investigate whether a short-term culture of tumour fragments irradiated ex vivo could anticipate patient responses to chemo- and/or radiotherapies. Biopsies were collected prior to treatment from a cohort of 28 patients with non-operable tumours of the oral cavity or oropharynx, and then cultured ex vivo. Short-term biopsy slice culture is a robust method that keeps cells viable for 7 days. Different biomarkers involved in the stemness status (CD44) or the DNA damage response (pATM and γ-H2AX) were investigated for their potential to predict the treatment response. A higher expression of all these markers was predictive of a poor response to treatment. This allowed the stratification of responder or non-responder patients to treatment. Moreover, the ratio for the expression of the three markers 24 h after 4 Gy irradiation versus 0 Gy was higher in responder than in non-responder patients. Finally, combining these biomarkers greatly improved their predictive potential, especially when the γ-H2AX ratio was associated with the CD44 ratio or the pATM ratio. These results encourage further evaluation of these biomarkers in a larger cohort of patients.
Collapse
|
3
|
Rassamegevanon T, Feindt L, Koi L, Müller J, Freudenberg R, Löck S, Sihver W, Çevik E, Kühn AC, von Neubeck C, Linge A, Pietzsch HJ, Kotzerke J, Baumann M, Krause M, Dietrich A. Molecular Response to Combined Molecular- and External Radiotherapy in Head and Neck Squamous Cell Carcinoma (HNSCC). Cancers (Basel) 2021; 13:cancers13225595. [PMID: 34830750 PMCID: PMC8615625 DOI: 10.3390/cancers13225595] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/03/2021] [Indexed: 01/11/2023] Open
Abstract
Simple Summary Our previous preclinical trial in a head and neck squamous cell carcinoma (HNSCC) xenograft model showed a high potential for the improvement of curative treatment outcome upon the combination treatment of a radiolabeled (Yttrium-90) anti-EGFR antibody (Cetuximab) and external radiotherapy. We aim to elucidate the molecular response of HNSCC tumors upon this combination. Here, we show that the combination treatment leads to an increasing number and complexity of DNA double strand breaks. The upregulation of p21cip1/waf1 expression and cleaved caspase-3 suggest a blockage of cell cycle transition and an induction of programmed cell death. Collectively, a complex interplay between molecular mechanisms involved in cell death induction, cell cycle arrest, and DNA double strand break repair accounts for the beneficial potential using Yttrium-90-Cetuximab in combination with external radiotherapy. Abstract Combination treatment of molecular targeted and external radiotherapy is a promising strategy and was shown to improve local tumor control in a HNSCC xenograft model. To enhance the therapeutic value of this approach, this study investigated the underlying molecular response. Subcutaneous HNSCC FaDuDD xenografts were treated with single or combination therapy (X-ray: 0, 2, 4 Gy; anti-EGFR antibody (Cetuximab) (un-)labeled with Yttrium-90 (90Y)). Tumors were excised 24 h post respective treatment. Residual DNA double strand breaks (DSB), mRNA expression of DNA damage response related genes, immunoblotting, tumor histology, and immunohistological staining were analyzed. An increase in number and complexity of residual DNA DSB was observed in FaDuDD tumors exposed to the combination treatment of external irradiation and 90Y-Cetuximab relative to controls. The increase was observed in a low oxygenated area, suggesting the expansion of DNA DSB damages. Upregulation of genes encoding p21cip1/waf1 (CDKN1A) and GADD45α (GADD45A) was determined in the combination treatment group, and immunoblotting as well as immunohistochemistry confirmed the upregulation of p21cip1/waf1. The increase in residual γH2AX foci leads to the blockage of cell cycle transition and subsequently to cell death, which could be observed in the upregulation of p21cip1/waf1 expression and an elevated number of cleaved caspase-3 positive cells. Overall, a complex interplay between DNA damage repair and programmed cell death accounts for the potential benefit of the combination therapy using 90Y-Cetuximab and external radiotherapy.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
| | - Louis Feindt
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Lydia Koi
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Johannes Müller
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | - Robert Freudenberg
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.F.); (J.K.)
| | - Steffen Löck
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Wiebke Sihver
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (W.S.); (H.-J.P.)
| | - Enes Çevik
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
- School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Ariane Christel Kühn
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- B CUBE—Center for Molecular Bioengineering, Technische Universität Dresden, 01307 Dresden, Germany
| | - Cläre von Neubeck
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Particle Therapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany
| | - Annett Linge
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Hans-Jürgen Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, 01328 Dresden, Germany; (W.S.); (H.-J.P.)
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (R.F.); (J.K.)
| | - Michael Baumann
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
| | - Mechthild Krause
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, 01307 Dresden, Germany
| | - Antje Dietrich
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany; (T.R.); (S.L.); (C.v.N.); (A.L.); (M.K.)
- German Cancer Research Center (DKFZ), 69192 Heidelberg, Germany;
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, 01307 Dresden, Germany; (L.F.); (L.K.); (J.M.); (E.Ç.); (A.C.K.)
- Correspondence: ; Tel.: +49-351-458-7404
| |
Collapse
|
4
|
Grgic I, Tschanz F, Borgeaud N, Gupta A, Clavien PA, Guckenberger M, Graf R, Pruschy M. Tumor Oxygenation by Myo-Inositol Trispyrophosphate Enhances Radiation Response. Int J Radiat Oncol Biol Phys 2021; 110:1222-1233. [PMID: 33587991 DOI: 10.1016/j.ijrobp.2021.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 01/18/2021] [Accepted: 02/07/2021] [Indexed: 12/29/2022]
Abstract
PURPOSE Tumor hypoxia is a major limiting factor for successful radiation therapy outcomes, with hypoxic cells being up to 3-fold more radiation resistant than normoxic cells; tumor hypoxia creates a tumor microenvironment that is hostile to immune response. Thus, pharmaceutical-induced tumor oxygenation before radiation therapy represents an interesting method to enhance the efficacy of radiation therapy. Myo-inositol trispyrophosphate (ITPP) triggers a decrease in the affinity of oxygen to hemoglobin, which leads to an increased release of oxygen upon tissue demand, including in hypoxic tumors. METHODS AND MATERIALS The combined treatment modality of high-dose bolus ITPP with a single high-dose fraction of ionizing radiation (IR) was investigated for its mechanics and efficacy in multiple preclinical animal tumor models in immunocompromised and immunocompetent mice. The dynamics of tumor oxygenation were determined by serial hypoxia-oriented bioimaging. Initial and residual DNA damage and the integrity of the tumor vasculature were quantified on the immunohistochemical level in response to the different treatment combinations. RESULTS ITPP application did not affect tumor growth as a single treatment modality, but it rapidly induced tumor oxygenation, as demonstrated by in vivo imaging, and significantly reduced tumor growth when combined with IR. An immunohistochemical analysis of γH2AX foci demonstrated increased initial and residual IR-induced DNA damage as the primary mechanism for radiosensitization within initially hypoxic but ITPP-oxygenated tumor regions. Scheduling experiments revealed that ITPP increases the efficacy of ionizing radiation only when applied before radiation therapy. Irradiation alone damaged the tumor vasculature and increased tumor hypoxia, which were both prevented by combined treatment with ITPP. Interestingly, the combined treatment modality also promoted increased immune cell infiltration. CONCLUSIONS ITPP-mediated tumor oxygenation and vascular protection triggers immediate and delayed processes to enhance the efficacy of ionizing radiation for successful radiation therapy.
Collapse
Affiliation(s)
- Ivo Grgic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University Zurich, Zurich, Switzerland
| | - Fabienne Tschanz
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University Zurich, Zurich, Switzerland
| | - Nathalie Borgeaud
- Laboratory of the Swiss-Hepato-Pancreatico-Biliary (HPB) Centre, Department of Visceral Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Anurag Gupta
- Laboratory of the Swiss-Hepato-Pancreatico-Biliary (HPB) Centre, Department of Visceral Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Pierre-Alain Clavien
- Laboratory of the Swiss-Hepato-Pancreatico-Biliary (HPB) Centre, Department of Visceral Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University Zurich, Zurich, Switzerland
| | - Rolf Graf
- Laboratory of the Swiss-Hepato-Pancreatico-Biliary (HPB) Centre, Department of Visceral Surgery, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Riedel A, Klumpp L, Menegakis A, De-Colle C, Huber SM, Schittenhelm J, Neumann M, Noell S, Tatagiba M, Zips D. γH2AX foci assay in glioblastoma: Surgical specimen versus corresponding stem cell culture. Radiother Oncol 2021; 159:119-125. [PMID: 33775712 DOI: 10.1016/j.radonc.2021.03.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/02/2020] [Accepted: 03/17/2021] [Indexed: 11/29/2022]
Abstract
AIM To assess radiation response using γH2AX assay in surgical specimens from glioblastoma (GB) patients and their corresponding primary gliosphere culture. To test the hypothesis that gliospheres (stem cell enriched) are more resistant than specimens (bulky cell dominated) but that the interpatient heterogeneity is similar. MATERIAL AND METHODS Ten pairs of specimens and corresponding gliospheres derived from patients with IDH-wildtype GB were studied. Specimens and gliospheres were irradiated with graded doses and after 24 h the number of residual γH2AX foci was counted. RESULTS Gliospheres showed a higher Nestin expression than specimens and exhibited two different phenotypes: free floating (n = 7) and attached (n = 3). Slope analysis revealed an interpatient heterogeneity with values between 0.15 and 1.30 residual γH2AX foci/Gy. Free-floating spheres were more resistant than their parental specimens (median slope 0.13 foci/Gy versus 0.53) as well as than the attached spheres (2.14). The slopes of free floating spheres did not correlate with their corresponding specimens while a trend for a positive correlation was found for the attached spheres and the respective specimens. Association with MGMT did not reach statistical significance. CONCLUSION Consistent with the clinical phenotype and our previous experiments, GB specimens show low radiation sensitivity. Stem-cell enriched free-floating gliospheres were more resistant than specimens supporting the concept of radioresistance in stem cell-like cells. The lack of correlation between specimens and their respective gliosphere cultures needs validation and may have a profound impact on future translational studies using γH2AX as a potential biomarker for personalized radiation therapy.
Collapse
Affiliation(s)
- Andreas Riedel
- Radiation Oncology, Medical Faculty and University Hospital Tübingen, Germany
| | - Lukas Klumpp
- Radiation Oncology, Medical Faculty and University Hospital Tübingen, Germany
| | - Apostolos Menegakis
- Netherlands Cancer Institute, Division of Cell Biology, Amsterdam, The Netherlands
| | - Chiara De-Colle
- Radiation Oncology, Medical Faculty and University Hospital Tübingen, Germany
| | - Stephan M Huber
- Radiation Oncology, Medical Faculty and University Hospital Tübingen, Germany
| | - Jens Schittenhelm
- Division of Neuropathology, Medical Faculty and University Hospital Tübingen, Germany
| | - Manuela Neumann
- Division of Neuropathology, Medical Faculty and University Hospital Tübingen, Germany
| | - Susan Noell
- Department of Neurosurgery, Medical Faculty and University Hospital Tübingen, Germany
| | - Marcos Tatagiba
- Department of Neurosurgery, Medical Faculty and University Hospital Tübingen, Germany
| | - Daniel Zips
- Radiation Oncology, Medical Faculty and University Hospital Tübingen, Germany; German Cancer Consortium (DKTK), Partner Site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
Resistance of Hypoxic Cells to Ionizing Radiation Is Mediated in Part via Hypoxia-Induced Quiescence. Cells 2021; 10:cells10030610. [PMID: 33801903 PMCID: PMC7998378 DOI: 10.3390/cells10030610] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Double strand breaks (DSBs) are highly toxic to a cell, a property that is exploited in radiation therapy. A critical component for the damage induction is cellular oxygen, making hypoxic tumor areas refractory to the efficacy of radiation treatment. During a fractionated radiation regimen, these hypoxic areas can be re-oxygenated. Nonetheless, hypoxia still constitutes a negative prognostic factor for the patient’s outcome. We hypothesized that this might be attributed to specific hypoxia-induced cellular traits that are maintained upon reoxygenation. Here, we show that reoxygenation of hypoxic non-transformed RPE-1 cells fully restored induction of DSBs but the cells remain radioresistant as a consequence of hypoxia-induced quiescence. With the use of the cell cycle indicators (FUCCI), cell cycle-specific radiation sensitivity, the cell cycle phase duration with live cell imaging, and single cell tracing were assessed. We observed that RPE-1 cells experience a longer G1 phase under hypoxia and retain a large fraction of cells that are non-cycling. Expression of HPV oncoprotein E7 prevents hypoxia-induced quiescence and abolishes the radioprotective effect. In line with this, HPV-negative cancer cell lines retain radioresistance, while HPV-positive cancer cell lines are radiosensitized upon reoxygenation. Quiescence induction in hypoxia and its HPV-driven prevention was observed in 3D multicellular spheroids. Collectively, we identify a new hypoxia-dependent radioprotective phenotype due to hypoxia-induced quiescence that accounts for a global decrease in radiosensitivity that can be retained upon reoxygenation and is absent in cells expressing oncoprotein E7.
Collapse
|
7
|
Rassamegevanon T, Löck S, Baumann M, Krause M, von Neubeck C. Comparable radiation response of ex vivo and in vivo irradiated tumor samples determined by residual γH2AX. Radiother Oncol 2019; 139:94-100. [PMID: 31445839 DOI: 10.1016/j.radonc.2019.06.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/16/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE a) To investigate if an ex vivo cultured and irradiated tumor biopsy reflects and predicts the radiation response of the corresponding in vivo irradiated tumor measured with the DNA double strand break marker γH2AX foci. MATERIALS AND METHODS Five human head and neck squamous cell carcinoma (hHNSCC) xenograft models were used. Fine needle biopsies were taken from anesthetized tumor-bearing NMRI nude mice prior to in vivo single dose irradiation (0, 2, 4, or 8 Gy) under ambient blood flow. Biopsies were ex vivo reoxygenated and irradiated with equivalent doses. Tumors and biopsies were fixed 24 h post irradiation, and γH2AX foci were assessed in oxygenated tumor regions. RESULTS Linear regression analysis showed comparable slopes of the residual γH2AX foci dose-response curves in four out of five hHNSCC models when in vivo and ex vivo cohorts were compared. The slopes from ex vivo biopsies and in vivo tumors could classify the respective tumor model as sensitive or resistant according to the intrinsic radiation sensitivity (TCD50). CONCLUSION The ability of ex vivo irradiated tumor biopsies to reflect and predict the intrinsic radiation response of in vivo tumors increases the translational potential of the ex vivo γH2AX foci assay as a diagnostic tool for clinical practice.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany.
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany; National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, and; Helmholtz Association / Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Cläre von Neubeck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
8
|
Meneceur S, Löck S, Gudziol V, Hering S, Bütof R, Rehm M, Baumann M, Krause M, von Neubeck C. Residual gammaH2AX foci in head and neck squamous cell carcinomas as predictors for tumour radiosensitivity: Evaluation in pre-clinical xenograft models and clinical specimens. Radiother Oncol 2019; 137:24-31. [DOI: 10.1016/j.radonc.2019.04.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
|
9
|
Rassamegevanon T, Löck S, Baumann M, Krause M, von Neubeck C. Heterogeneity of γH2AX Foci Increases in Ex Vivo Biopsies Relative to In Vivo Tumors. Int J Mol Sci 2018; 19:E2616. [PMID: 30181446 PMCID: PMC6163410 DOI: 10.3390/ijms19092616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/30/2022] Open
Abstract
The biomarker for DNA double stand breaks, gammaH2AX (γH2AX), holds a high potential as an intrinsic radiosensitivity predictor of tumors in clinical practice. Here, two published γH2AX foci datasets from in and ex vivo exposed human head and neck squamous cell carcinoma (hHNSCC) xenografts were statistically re-evaluated for the effect of the assay setting (in or ex vivo) on cellular geometry and the degree of heterogeneity in γH2AX foci. Significant differences between the nucleus areas of in- and ex vivo exposed samples were found. However, the number of foci increased linearly with nucleus area in irradiated samples of both settings. Moreover, irradiated tumor cells showed changes of nucleus area distributions towards larger areas compared to unexposed samples, implying cell cycle alteration after radiation exposure. The number of residual γH2AX foci showed a higher degree of intra-tumoral heterogeneity in the ex vivo exposed samples relative to the in vivo exposed samples. In the in vivo setting, the highest intra-tumoral heterogeneity was observed in initial γH2AX foci numbers (foci detected 30 min following irradiation). These results suggest that the tumor microenvironment and the culture condition considerably influence cellular adaptation and DNA damage repair.
Collapse
Affiliation(s)
- Treewut Rassamegevanon
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Steffen Löck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Michael Baumann
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Mechthild Krause
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany.
| | - Cläre von Neubeck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
10
|
Klumpp L, Sezgin EC, Skardelly M, Eckert F, Huber SM. KCa3.1 Channels and Glioblastoma: In Vitro Studies. Curr Neuropharmacol 2018; 16:627-635. [PMID: 28786347 PMCID: PMC5997865 DOI: 10.2174/1570159x15666170808115821] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 06/29/2017] [Accepted: 07/12/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Several tumor entities including brain tumors aberrantly overexpress intermediate conductance Ca2+ activated KCa3.1 K+ channels. These channels contribute significantly to the transformed phenotype of the tumor cells. METHOD PubMed was searched in order to summarize our current knowledge on the molecular signaling upstream and downstream and the effector functions of KCa3.1 channel activity in tumor cells in general and in glioblastoma cells in particular. In addition, KCa3.1 expression and function for repair of DNA double strand breaks was determined experimentally in primary glioblastoma cultures in dependence on the abundance of proneural and mesenchymal stem cell markers. RESULTS By modulating membrane potential, cell volume, Ca2+ signals and the respiratory chain, KCa3.1 channels in both, plasma and inner mitochondrial membrane, have been demonstrated to regulate many cellular processes such as migration and tissue invasion, metastasis, cell cycle progression, oxygen consumption and metabolism, DNA damage response and cell death of cancer cells. Moreover, KCa3.1 channels have been shown to crucially contribute to resistance against radiotherapy. Futhermore, the original in vitro data on KCa3.1 channel expression in subtypes of glioblastoma stem(-like) cells propose KCa3.1 as marker for the mesenchymal subgroup of cancer stem cells and suggest that KCa3.1 contributes to the therapy resistance of mesenchymal glioblastoma stem cells. CONCLUSION The data suggest KCa3.1 channel targeting in combination with radiotherapy as promising new tool to eradicate therapy-resistant mesenchymal glioblastoma stem cells.
Collapse
Affiliation(s)
| | | | | | | | - Stephan M. Huber
- Address correspondence to this author at the Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; Tel: +49-(0)7071-29-82183; E-mail:
| |
Collapse
|
11
|
Ex vivo γH2AX radiation sensitivity assay in prostate cancer: Inter-patient and intra-patient heterogeneity. Radiother Oncol 2017; 124:386-394. [PMID: 28919005 DOI: 10.1016/j.radonc.2017.08.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/14/2017] [Accepted: 08/25/2017] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The aim of the study is to assess inter-patient and intra-patient heterogeneity in tumour cell radiosensitivity using the ex vivo γH2AX assay in prostate cancer specimens. METHODS Excised specimens from untreated prostate cancer patients were cultivated 24h in media, irradiated ex vivo and fixed after 24h. Residual γH2AX foci were counted and the slope of the dose response was calculated. Intra-patient heterogeneity was studied from three to seven different biopsies. RESULTS In pathology-confirmed tumour samples from 21 patients the slope of residual γH2AX foci and radiation dose showed a substantial heterogeneity ranging from 0.82 to 3.17 foci/Gy. No correlation was observed between the slope values and the Gleason score (p=0.37), prostate specific antigen (p=0.48) and tumour stage (p=0.89). ANOVA indicated that only in 1 out of 9 patients, biopsies from different tumour locations yielded statistically significant differences. Variance component analysis indicated higher inter-patient than intra-patient variability. Bootstrap simulation study demonstrated that one biopsy is sufficient to estimate the mean value of residual γH2AX per dose level and account for intra-patient heterogeneity. CONCLUSIONS In prostate cancer inter-patient heterogeneity in tumour cell radiation sensitivity is pronounced and higher than intra-patient heterogeneity supporting the further development of the γH2AX ex vivo assay as a biomarker for individualized treatment.
Collapse
|
12
|
Tumor heterogeneity determined with a γH2AX foci assay: A study in human head and neck squamous cell carcinoma (hHNSCC) models. Radiother Oncol 2017; 124:379-385. [PMID: 28739384 DOI: 10.1016/j.radonc.2017.06.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/26/2017] [Accepted: 06/28/2017] [Indexed: 11/20/2022]
Abstract
PURPOSE This study aimed to analyze the intra-tumoral heterogeneity of γH2AX foci in tumor specimens following ex vivo radiation to evaluate the potential of γH2AX foci as predictors for radiosensitivity. MATERIAL AND METHODS γH2AX foci were quantified in tumor specimens of 3hHNSCC tumor models with known differences in radiosensitivity after reoxygenation in culture medium (10h, 24h), single dose exposure (0Gy, 4Gy), and fixation 24h post-irradiation. Multiple, equally treated samples of the same tumor were analyzed for foci, normalized and fitted in a linear mixed-effects model. RESULTS The ex vivo reoxygenation time had no significant effect on γH2AX foci counts. A significant intra model heterogeneity could be shown for FaDu (p=0.033) but not for SKX (p=0.167) and UT-SCC-5 (p=0.082) tumors, respectively. All tumor models showed a significant intra-tumoral heterogeneity between specimens of the same tumor (p<0.01) or among microscopic fields of a particular tumor specimen (p<0.0001). CONCLUSION Similar results for ex vivo γH2AX foci between 10h and 24h reoxygenation time support the applicability of the assay in a clinical setting. The high intra-tumoral heterogeneity underlines the necessity of multiple analyzable samples per patient and therewith the need for an automated foci analysis.
Collapse
|
13
|
Hauth F, Toulany M, Zips D, Menegakis A. Cell-line dependent effects of hypoxia prior to irradiation in squamous cell carcinoma lines. Clin Transl Radiat Oncol 2017; 5:12-19. [PMID: 29594212 PMCID: PMC5833923 DOI: 10.1016/j.ctro.2017.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/25/2017] [Accepted: 06/02/2017] [Indexed: 02/07/2023] Open
Abstract
Purpose To assess the impact of hypoxia exposure on cellular radiation sensitivity and survival of tumor cells with diverse intrinsic radiation sensitivity under normoxic conditions. Materials and methods Three squamous cell carcinoma (SCC) cell lines, with pronounced differences in radiation sensitivity, were exposed to hypoxia prior, during or post irradiation. Cells were seeded in parallel for colony formation assay (CFA) and stained for γH2AX foci or processed for western blot analysis. Results Hypoxia during irradiation led to increased cellular survival and reduced amount of residual γH2AX foci in all the cell lines with similar oxygen enhancement ratios (OER SKX: 2.31, FaDu: 2.44, UT-SCC5: 2.32), while post-irradiation hypoxia did not alter CFA nor residual γH2AX foci. Interestingly, prolonged exposure to hypoxia prior to irradiation resulted in differential outcome, assessed as Hypoxia modifying factor (HMF) namely radiosensitization (SKX HMF: 0.76), radioresistance (FaDu HMF: 1.54) and no effect (UT SCC-5 HMF: 1.1). Notably, radiosensitization was observed in the ATM-deficient SKX cell line while UT SCC-5 and to a lesser extent also FaDu cells showed radiation- and hypoxia-induced upregulation of ATM phosphorylation. Across all the cell lines Rad51 was downregulated whereas phosphor-DNA-PKcs was enhanced under hypoxia for FaDu and UTSCC-5 and was delayed in the SKX cell line. Conclusion We herein report a key role of ATM in the cellular fitness of cells exposed to prolonged moderate hypoxia prior to irradiation. While DNA damage response post-irradiation seem to be mainly driven by non-homologous end joining repair pathway in these conditions, our data suggest an important role for ATM kinase in hypoxia-driven modification of radiation response.
Collapse
Affiliation(s)
- Franziska Hauth
- Division of Radiobiology & Molecular Environmental Research Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Mahmoud Toulany
- Division of Radiobiology & Molecular Environmental Research Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Division of Radiobiology & Molecular Environmental Research Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Consortium for Translational Cancer Research (DKTK) Partner Sites Tübingen, Germany
| | - Apostolos Menegakis
- Division of Radiobiology & Molecular Environmental Research Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
- Corresponding author at: Department of Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Hoppe-Seylerstrasse 3, 72076 Tuebingen, Germany.Department of Radiation OncologyMedical Faculty and University HospitalEberhard Karls University TübingenHoppe-Seylerstrasse 372076 TuebingenGermany
| |
Collapse
|
14
|
Dynamic In Vivo Profiling of DNA Damage and Repair after Radiotherapy Using Canine Patients as a Model. Int J Mol Sci 2017; 18:ijms18061176. [PMID: 28587165 PMCID: PMC5485999 DOI: 10.3390/ijms18061176] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 01/22/2023] Open
Abstract
Time resolved data of DNA damage and repair after radiotherapy elucidates the relation between damage, repair, and cell survival. While well characterized in vitro, little is known about the time-course of DNA damage response in tumors sampled from individual patients. Kinetics of DNA damage after radiotherapy was assessed in eight dogs using repeated in vivo samples of tumor and co-irradiated normal tissue analyzed with comet assay and phosphorylated H2AX (γH2AX) immunohistochemistry. In vivo results were then compared (in silico) with a dynamic mathematical model for DNA damage formation and repair. Maximum %DNA in tail was observed at 15–60 min after irradiation, with a rapid decrease. Time-courses of γH2AX-foci paralleled these findings with a small time delay and were not influenced by covariates. The evolutionary parameter search based on %DNA in tail revealed a good fit of the DNA repair model to in vivo data for pooled sarcoma time-courses, but fits for individual sarcoma time-courses suffer from the heterogeneous nature of the in vivo data. It was possible to follow dynamics of comet tail intensity and γH2AX-foci during a course of radiation using a minimally invasive approach. DNA repair can be quantitatively investigated as time-courses of individual patients by integrating this resulting data into a dynamic mathematical model.
Collapse
|
15
|
Yamamoto VN, Thylur DS, Bauschard M, Schmale I, Sinha UK. Overcoming radioresistance in head and neck squamous cell carcinoma. Oral Oncol 2016; 63:44-51. [PMID: 27938999 DOI: 10.1016/j.oraloncology.2016.11.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 08/29/2016] [Accepted: 11/06/2016] [Indexed: 12/28/2022]
Abstract
Radiation therapy plays an essential role in the treatment of head and neck squamous cell carcinoma (HNSCC), yet therapeutic efficacy is hindered by treatment-associated toxicity and tumor recurrence. In comparison to other cancers, innovation has proved challenging, with the epidermal growth factor receptor (EGFR) antibody cetuximab being the only new radiosensitizing agent approved by the FDA in over half a century. This review examines the physiological mechanisms that contribute to radioresistance in HNSCC as well as preclinical and clinical data regarding novel radiosensitizing agents, with an emphasis on those with highest translational promise.
Collapse
Affiliation(s)
- Vicky N Yamamoto
- USC Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.
| | - David S Thylur
- USC Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Michael Bauschard
- USC Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Isaac Schmale
- Department of Otolaryngology-Head & Neck Surgery, University of Rochester Medical Center, Rochester, NY, United States
| | - Uttam K Sinha
- USC Tina and Rick Caruso Department of Otolaryngology-Head & Neck Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
16
|
Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, Richter C, Zips D, Bortfeld T. Radiation oncology in the era of precision medicine. Nat Rev Cancer 2016; 16:234-49. [PMID: 27009394 DOI: 10.1038/nrc.2016.18] [Citation(s) in RCA: 525] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Technological advances and clinical research over the past few decades have given radiation oncologists the capability to personalize treatments for accurate delivery of radiation dose based on clinical parameters and anatomical information. Eradication of gross and microscopic tumours with preservation of health-related quality of life can be achieved in many patients. Two major strategies, acting synergistically, will enable further widening of the therapeutic window of radiation oncology in the era of precision medicine: technology-driven improvement of treatment conformity, including advanced image guidance and particle therapy, and novel biological concepts for personalized treatment, including biomarker-guided prescription, combined treatment modalities and adaptation of treatment during its course.
Collapse
Affiliation(s)
- Michael Baumann
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Oncology, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Mechthild Krause
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Oncology, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C, Denmark
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120 Heidelberg
- Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 400, 69120 Heidelberg
- German Cancer Consortium (DKTK) Heidelberg, Germany
| | - Søren M Bentzen
- Department of Epidemiology and Public Health and Greenebaum Cancer Center, University of Maryland School of Medicine, 22 S Greene Street S9a03, Baltimore, Maryland 21201, USA
| | - Juliane Daartz
- Department of Radiation Oncology, Physics Division, Massachusetts General Hospital and Harvard Medical School, 1000 Blossom Street Cox 362, Boston, Massachusetts 02114, USA
| | - Christian Richter
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Daniel Zips
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Consortium Tübingen, Postfach 2669, 72016 Tübingen
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, Hoppe-Seyler-Strasse 3, 72016 Tübingen, Germany
| | - Thomas Bortfeld
- Department of Radiation Oncology, Physics Division, Massachusetts General Hospital and Harvard Medical School, 1000 Blossom Street Cox 362, Boston, Massachusetts 02114, USA
| |
Collapse
|
17
|
Tillner F, Thute P, Löck S, Dietrich A, Fursov A, Haase R, Lukas M, Rimarzig B, Sobiella M, Krause M, Baumann M, Bütof R, Enghardt W. Precise image-guided irradiation of small animals: a flexible non-profit platform. Phys Med Biol 2016; 61:3084-108. [DOI: 10.1088/0031-9155/61/8/3084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
18
|
Fliedner FP, Hansen AE, Jørgensen JT, Kjær A. The use of matrigel has no influence on tumor development or PET imaging in FaDu human head and neck cancer xenografts. BMC Med Imaging 2016; 16:5. [PMID: 26762341 PMCID: PMC4712613 DOI: 10.1186/s12880-016-0105-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/04/2016] [Indexed: 11/10/2022] Open
Abstract
Background In preclinical research MatrixgelTM Basement Membrane Matrix (MG) is used frequently for the establishment of syngeneic and xenograft cancer models. Limited information on its influence on parameters including; tumor growth, vascularization, hypoxia and imaging characteristics is currently available. This study evaluates the potential effect of matrigel use in a human head and neck cancer xenograft model (FaDu; hypopharyngeal carcinoma) in NMRI nude mice. The FaDu cell line was chosen based on its frequent use in studies of cancer imaging and tumor microenvironment. Methods NMRI nude mice (n = 34) were divided into two groups and subcutaneously injected with FaDu cells in medium either including (+MG) or excluding matrigel (−MG). In sub study I seven mice from each group (+MG, n = 7; −MG, n = 7) were 18F- fluorodeoxyglucose (18F-FDG) PET/CT scanned on Day 5, 8, 12, 15, and 19. In sub study II ten mice from each group (+MG, n = 10; −MG, n = 10) were included and tumors collected for immunohistochemistry (IHC) analysis of tumor microenvironment including; proliferation ratio, micro vessel density, average vessel area, hypoxia, nuclear density, and necrosis. Tumors for IHC were collected according to size (200–400 mm3, 500–700 mm3, 800–1100 mm3). Results FDG uptake and tumor growth was statistically compatible for the tumors established with or without MG. The IHC analysis on all parameters only identified a significantly higher micro vessel density for tumor size 500–700 mm3 and 800–1100 mm3 and average vessel area for tumor size 500–700 mm3 in the −MG group. Comparable variations were observed for tumors of both the +MG and −MG groups. No difference in tumor take rate was observed between groups in study. Conclusions Matrigel did not affect tumor growth or tumor take for the FaDu xenograft model evaluated. Tumors in the -MG group displayed increased angiogenesis compared to the +MG tumors. No difference in 18F-FDG PET uptake for tumors of different groups was found. Based on these observations the influence of matrigel on tumor imaging and tumor microenvironment seems minor for this particular xenograft model.
Collapse
Affiliation(s)
- Frederikke P Fliedner
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Anders E Hansen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark. .,Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
19
|
Bell C, Dowson N, Fay M, Thomas P, Puttick S, Gal Y, Rose S. Hypoxia imaging in gliomas with 18F-fluoromisonidazole PET: toward clinical translation. Semin Nucl Med 2015; 45:136-50. [PMID: 25704386 DOI: 10.1053/j.semnuclmed.2014.10.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
There is significant interest in the development of improved image-guided therapy for neuro-oncology applications. Glioblastomas (GBM) in particular present a considerable challenge because of their pervasive nature, propensity for recurrence, and resistance to conventional therapies. MRI is routinely used as a guide for planning treatment strategies. However, this imaging modality is not able to provide images that clearly delineate tumor boundaries and affords only indirect information about key tumor pathophysiology. With the emergence of PET imaging with new oncology radiotracers, mapping of tumor infiltration and other important molecular events such as hypoxia is now feasible within the clinical setting. In particular, the importance of imaging hypoxia levels within the tumoral microenvironment is gathering interest, as hypoxia is known to play a central role in glioma pathogenesis and resistance to treatment. One of the hypoxia radiotracers known for its clinical utility is (18)F-fluoromisodazole ((18)F-FMISO). In this review, we highlight the typical causes of treatment failure in gliomas that may be linked to hypoxia and outline current methods for the detection of hypoxia. We also provide an overview of the growing body of studies focusing on the clinical translation of (18)F-FMISO PET imaging, strengthening the argument for the use of (18)F-FMISO hypoxia imaging to help optimize and guide treatment strategies for patients with glioblastoma.
Collapse
Affiliation(s)
- Christopher Bell
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; School of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Nicholas Dowson
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Mike Fay
- Department of Radiation Oncology, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
| | - Paul Thomas
- Specialised PET Services Queensland, Royal Brisbane and Women's Hospital, Herston, Brisbane, Queensland, Australia
| | - Simon Puttick
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Yaniv Gal
- Centre for Medical Diagnostic Technologies in Queensland, University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Stephen Rose
- CSIRO Preventative Health Flagship, CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; CSIRO Computational Informatics, The Australian e-Health Research Centre, Herston, Queensland, Australia; School of Medicine, University of Queensland, St Lucia, Brisbane, Queensland, Australia.
| |
Collapse
|
20
|
Tinhofer I, Niehr F, Konschak R, Liebs S, Munz M, Stenzinger A, Weichert W, Keilholz U, Budach V. Next-generation sequencing: hype and hope for development of personalized radiation therapy? Radiat Oncol 2015; 10:183. [PMID: 26316159 PMCID: PMC4554356 DOI: 10.1186/s13014-015-0481-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/07/2015] [Indexed: 01/18/2023] Open
Abstract
The introduction of next-generation sequencing (NGS) in the field of cancer research has boosted worldwide efforts of genome-wide personalized oncology aiming at identifying predictive biomarkers and novel actionable targets. Despite considerable progress in understanding the molecular biology of distinct cancer entities by the use of this revolutionary technology and despite contemporaneous innovations in drug development, translation of NGS findings into improved concepts for cancer treatment remains a challenge. The aim of this article is to describe shortly the NGS platforms for DNA sequencing and in more detail key achievements and unresolved hurdles. A special focus will be given on potential clinical applications of this innovative technique in the field of radiation oncology.
Collapse
Affiliation(s)
- Ingeborg Tinhofer
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany. .,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany.
| | - Franziska Niehr
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Robert Konschak
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Sandra Liebs
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| | - Matthias Munz
- Group for Computational Modelling in Medicine, Institute for Theoretical Biology, Humboldt Universität, Berlin, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany.,Institute of Pathology, University Hospital and National Center for Tumor Diseases, Heidelberg, Germany
| | - Ulrich Keilholz
- Charité Comprehensive Cancer Center, Charité University Hospital, Berlin, Germany
| | - Volker Budach
- Department of Radiooncology and Radiotherapy, Charité University Hospital Berlin, Translational Radiation Oncology Research Laboratory, Charitéplatz 1, 10117, Berlin, Germany.,German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK) partner site, Heidelberg, Germany
| |
Collapse
|
21
|
Menegakis A, De Colle C, Yaromina A, Hennenlotter J, Stenzl A, Scharpf M, Fend F, Noell S, Tatagiba M, Brucker S, Wallwiener D, Boeke S, Ricardi U, Baumann M, Zips D. Residual γH2AX foci after ex vivo irradiation of patient samples with known tumour-type specific differences in radio-responsiveness. Radiother Oncol 2015; 116:480-5. [PMID: 26297183 DOI: 10.1016/j.radonc.2015.08.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 08/04/2015] [Accepted: 08/06/2015] [Indexed: 02/01/2023]
Abstract
PURPOSE To apply our previously published residual ex vivo γH2AX foci method to patient-derived tumour specimens covering a spectrum of tumour-types with known differences in radiation response. In addition, the data were used to simulate different experimental scenarios to simplify the method. MATERIALS AND METHODS Evaluation of residual γH2AX foci in well-oxygenated tumour areas of ex vivo irradiated patient-derived tumour specimens with graded single doses was performed. Immediately after surgical resection, the samples were cultivated for 24h in culture medium prior to irradiation and fixed 24h post-irradiation for γH2AX foci evaluation. Specimens from a total of 25 patients (including 7 previously published) with 10 different tumour types were included. RESULTS Linear dose response of residual γH2AX foci was observed in all specimens with highly variable slopes among different tumour types ranging from 0.69 (95% CI: 1.14-0.24) to 3.26 (95% CI: 4.13-2.62) for chondrosarcomas (radioresistant) and classical seminomas (radiosensitive) respectively. Simulations suggest that omitting dose levels might simplify the assay without compromising robustness. CONCLUSION Here we confirm clinical feasibility of the assay. The slopes of the residual foci number are well in line with the expected differences in radio-responsiveness of different tumour types implying that intrinsic radiation sensitivity contributes to tumour radiation response. Thus, this assay has a promising potential for individualized radiation therapy and prospective validation is warranted.
Collapse
Affiliation(s)
- Apostolos Menegakis
- Department of Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Consortium for Translational Cancer Research (DKTK) Partner Sites Tübingen, Germany.
| | - Chiara De Colle
- Department of Oncology, Radiation Oncology, University of Turin, Italy
| | - Ala Yaromina
- Department of Radiation Oncology (Maastro), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, The Netherlands
| | - Joerg Hennenlotter
- Department of Urology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Arnulf Stenzl
- Department of Urology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Marcus Scharpf
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Falko Fend
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Susan Noell
- Department of Neurosurgery, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Marcos Tatagiba
- Department of Neurosurgery, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Sara Brucker
- Department of and Research Institute for Women's Health, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Diethelm Wallwiener
- Department of and Research Institute for Women's Health, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Simon Boeke
- Department of Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Consortium for Translational Cancer Research (DKTK) Partner Sites Tübingen, Germany
| | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, University of Turin, Italy
| | - Michael Baumann
- German Cancer Research Center (DKFZ), Heidelberg and German Consortium for Translational Cancer Research (DKTK) Partner Sites Dresden, Germany; Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Consortium for Translational Cancer Research (DKTK) Partner Sites Tübingen, Germany
| |
Collapse
|
22
|
Ree AH, Redalen KR. Personalized radiotherapy: concepts, biomarkers and trial design. Br J Radiol 2015; 88:20150009. [PMID: 25989697 DOI: 10.1259/bjr.20150009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In the past decade, and pointing onwards to the immediate future, clinical radiotherapy has undergone considerable developments, essentially including technological advances to sculpt radiation delivery, the demonstration of the benefit of adding concomitant cytotoxic agents to radiotherapy for a range of tumour types and, intriguingly, the increasing integration of targeted therapeutics for biological optimization of radiation effects. Recent molecular and imaging insights into radiobiology will provide a unique opportunity for rational patient treatment, enabling the parallel design of next-generation trials that formally examine the therapeutic outcome of adding targeted drugs to radiation, together with the critically important assessment of radiation volume and dose-limiting treatment toxicities. In considering the use of systemic agents with presumed radiosensitizing activity, this may also include the identification of molecular, metabolic and imaging markers of treatment response and tolerability, and will need particular attention on patient eligibility. In addition to providing an overview of clinical biomarker studies relevant for personalized radiotherapy, this communication will highlight principles in addressing clinical evaluation of combined-modality-targeted therapeutics and radiation. The increasing number of translational studies that bridge large-scale omics sciences with quality-assured phenomics end points-given the imperative development of open-source data repositories to allow investigators the access to the complex data sets-will enable radiation oncology to continue to position itself with the highest level of evidence within existing clinical practice.
Collapse
Affiliation(s)
- A H Ree
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway.,2 Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - K R Redalen
- 1 Department of Oncology, Akershus University Hospital, Lørenskog, Norway
| |
Collapse
|
23
|
Willers H, Gheorghiu L, Liu Q, Efstathiou JA, Wirth LJ, Krause M, von Neubeck C. DNA Damage Response Assessments in Human Tumor Samples Provide Functional Biomarkers of Radiosensitivity. Semin Radiat Oncol 2015; 25:237-50. [PMID: 26384272 DOI: 10.1016/j.semradonc.2015.05.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Predictive biomarkers are urgently needed for individualization of radiation therapy and treatment with radiosensitizing anticancer agents. Genomic profiling of human cancers provides us with unprecedented insight into the mutational landscape of genes directly or indirectly involved in the response to radiation-induced DNA damage. However, to what extent this wealth of structural information about the cancer genome produces biomarkers of sensitivity to radiation remains to be seen. Investigators are increasingly studying the subnuclear accumulation (ie, foci) of proteins in the DNA damage response (DDR), such as gamma-H2AX, 53BP1, or RAD51, as a surrogate of treatment sensitivity. Recent findings from preclinical studies have demonstrated the predictive potential of DDR foci by correlating foci with clinically relevant end points such as tumor control probability. Therefore, preclinical investigations of DDR foci responses are increasingly moving into cells and tissues from patients, which is the major focus of this review. The advantage of using DDR foci as functional biomarkers is that they can detect alterations in DNA repair due to various mechanisms. Moreover, they provide a global measurement of DDR network function without needing to know the identities of all the components, many of which remain unknown. Foci assays are thus expected to yield functional insight that may complement or supersede genomic information, thereby giving radiation oncologists unique opportunities to individualize cancer treatments in the near future.
Collapse
Affiliation(s)
- Henning Willers
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA.
| | - Liliana Gheorghiu
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Qi Liu
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Mechthild Krause
- German Cancer Consortium (DKTK) Dresden and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Germany
| | - Cläre von Neubeck
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| |
Collapse
|
24
|
γH2AX assay in ex vivo irradiated tumour specimens: A novel method to determine tumour radiation sensitivity in patient-derived material. Radiother Oncol 2015; 116:473-9. [PMID: 25866027 DOI: 10.1016/j.radonc.2015.03.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Revised: 02/25/2015] [Accepted: 03/23/2015] [Indexed: 11/21/2022]
Abstract
PURPOSE To establish a clinically applicable protocol for quantification of residual γH2AX foci in ex vivo irradiated tumour samples and to apply this method in a proof-of-concept feasibility study to patient-derived tumour specimens. MATERIAL AND METHODS Evaluation of γH2AX foci formation and disappearance in excised FaDu tumour specimens after (a) different incubation times in culture medium, 4Gy irradiation and fixation after 24h (cell recovery), (b) 10h medium incubation, 4Gy irradiation and fixation after various time points (double strand break repair kinetics), and (c) 10h medium incubation, irradiation with graded single radiation doses and fixation after 24h (dose-response). The optimised protocol was applied to patient-derived samples of seminoma, prostate cancer and glioblastoma multiforme. RESULTS Post excision or biopsy, tumour tissues showed stable radiation-induced γH2AX foci values in oxic cells after >6h of recovery in medium. Kinetics of foci disappearance indicated a plateau of residual foci after >12h following ex vivo irradiation. Fitting the dose-response of residual γH2AX foci yielded slopes comparable with in situ irradiation of FaDu tumours. Significant differences in the slopes of ex vivo irradiated patient-derived tumour samples were found. CONCLUSION A novel clinically applicable method to quantify residual γH2AX foci in ex vivo irradiated tumour samples was established. The first clinical results suggest that this method allows to distinguish between radiosensitive and radioresistant tumour types. These findings support further translational evaluation of this assay to individualise radiation therapy.
Collapse
|
25
|
Mäbert K, Cojoc M, Peitzsch C, Kurth I, Souchelnytskyi S, Dubrovska A. Cancer biomarker discovery: current status and future perspectives. Int J Radiat Biol 2014; 90:659-77. [PMID: 24524284 DOI: 10.3109/09553002.2014.892229] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE Cancer is a multigene disease which arises as a result of mutational and epigenetic changes coupled with activation of complex signaling networks. The use of biomarkers for early cancer detection, staging and individualization of therapy might improve patient care. A few fundamental issues such as tumor heterogeneity, a highly dynamic nature of the intrinsic and extrinsic determinants of radio- and chemoresistance, along with the plasticity and diversity of cancer stem cells (CSC) make biomarker development a challenging task. In this review we outline the preclinical strategies of cancer biomarker discovery including genomic, proteomic, metabolomic and microRNomic profiling, comparative genome hybridization (CGH), single nucleotide polymorphism (SNP) analysis, high throughput screening (HTS) and next generation sequencing (NGS). Other promising approaches such as assessment of circulating tumor cells (CTC), analysis of CSC-specific markers and cell-free circulating tumor DNA (ctDNA) are also discussed. CONCLUSIONS The emergence of powerful proteomic and genomic technologies in conjunction with advanced bioinformatic tools allows the simultaneous analysis of thousands of biological molecules. These techniques yield the discovery of new tumor signatures, which are sensitive and specific enough for early cancer detection, for monitoring disease progression and for proper treatment selection, paving the way to individualized cancer treatment.
Collapse
Affiliation(s)
- Katrin Mäbert
- OncoRay-National Center for Radiation Research in Oncology, Medical Faculty Dresden Carl Gustav Carus , TU Dresden , Germany
| | | | | | | | | | | |
Collapse
|
26
|
Economopoulou M, Masjkur J, Raiskup F, Ebermann D, Saha S, Karl MO, Funk R, Jaszai J, Chavakis T, Ehrhart-Bornstein M, Pillunat LE, Kunz-Schughart L, Kurth I, Dubrovska A, Androutsellis-Theotokis A. Expression of the transcription factor Hes3 in the mouse and human ocular surface, and in pterygium. Int J Radiat Biol 2014; 90:700-9. [DOI: 10.3109/09553002.2014.892228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
27
|
Pouliliou S, Koukourakis MI. Gamma histone 2AX (γ-H2AX)as a predictive tool in radiation oncology. Biomarkers 2014; 19:167-80. [DOI: 10.3109/1354750x.2014.898099] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Stamatia Pouliliou
- Department of Radiotherapy/Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace
AlexandroupolisGreece
| | - Michael I. Koukourakis
- Department of Radiotherapy/Oncology, Radiobiology and Radiopathology Unit, Democritus University of Thrace
AlexandroupolisGreece
| |
Collapse
|
28
|
Brüchner K, Beyreuther E, Baumann M, Krause M, Oppelt M, Pawelke J. Establishment of a small animal tumour model for in vivo studies with low energy laser accelerated particles. Radiat Oncol 2014; 9:57. [PMID: 24533586 PMCID: PMC3936820 DOI: 10.1186/1748-717x-9-57] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 02/10/2014] [Indexed: 12/12/2022] Open
Abstract
Background The long-term aim of developing a laser based acceleration of protons and ions towards clinical application requires not only substantial technological progress, but also the radiobiological characterization of the resulting ultra-short pulsed particle beams. Recent in vitro data showed similar effects of laser-accelerated versus "conventional" protons on clonogenic cell survival. As the proton energies currently achieved by laser driven acceleration are too low to penetrate standard tumour models on mouse legs, the aim of the present work was to establish a tumour model allowing for the penetration of low energy protons (~ 20 MeV) to further verify their effects in vivo. Methods KHT mouse sarcoma cells were injected subcutaneously in the right ear of NMRI (nu/nu) mice and the growing tumours were characterized with respect to growth parameters, histology and radiation response. In parallel, the laser system JETI was prepared for animal experimentation, i.e. a new irradiation setup was implemented and the laser parameters were carefully adjusted. Finally, a proof-of-principle experiment with laser accelerated electrons was performed to validate the tumour model under realistic conditions, i.e. altered environment and horizontal beam delivery. Results KHT sarcoma on mice ears showed a high take rate and continuous tumour growth after reaching a volume of ~ 5 mm3. The first irradiation experiment using laser accelerated electrons versus 200 kV X-rays was successfully performed and tumour growth delay was evaluated. Comparable tumour growth delay was found between X-ray and laser accelerated electron irradiation. Moreover, experimental influences, like anaesthesia and positioning at JETI, were found to be negligible. Conclusion A small animal tumour model suitable for the irradiation with low energy particles was established and validated at a laser based particle accelerator. Thus, the translation from in vitro to in vivo experimentation was for the first time realized allowing a broader preclinical validation of radiobiological characteristics and efficacy of laser driven particle accelerators in the future.
Collapse
Affiliation(s)
- Kerstin Brüchner
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, TU Dresden, Fetscherstr, 74, 01307 Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
29
|
Stegeman H, Span PN, Cockx SC, Peters JPW, Rijken PFJW, van der Kogel AJ, Kaanders JHAM, Bussink J. EGFR-Inhibition Enhances Apoptosis in Irradiated Human Head and Neck Xenograft Tumors Independent of Effects on DNA Repair. Radiat Res 2013; 180:414-21. [DOI: 10.1667/rr3349.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- H. Stegeman
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - P. N. Span
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - S. C. Cockx
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. P. W. Peters
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - P. F. J. W. Rijken
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - A. J. van der Kogel
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. H. A. M. Kaanders
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - J. Bussink
- Department of Radiation Oncology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Krause M, Kummer B, Deparade A, Eicheler W, Pfitzmann D, Yaromina A, Kunz-Schughart LA, Baumann M. Simultaneous PLK1 inhibition improves local tumour control after fractionated irradiation. Radiother Oncol 2013; 108:422-8. [PMID: 23891096 DOI: 10.1016/j.radonc.2013.06.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/24/2013] [Accepted: 06/28/2013] [Indexed: 12/30/2022]
Abstract
PURPOSE Polo-like kinase 1 (PLK1) plays an important role in mitotic progression, is frequently overexpressed and associated with a poor prognosis of cancer patients, thus providing a promising target in anticancer treatment. Aim of the current project was to evaluate the effect of the novel PLK1 inhibitor BI 6727 in combination with irradiation. MATERIAL AND METHODS In vitro proliferation and radiation cell survival assays as well as in vivo local tumour control assays after single treatment and combined radiation and drug application were carried out using the squamous cell carcinoma models A431 and FaDu. In addition, cell cycle phases were monitored in vitro and in vivo. RESULTS BI 6727 showed a dose-dependent antiproliferative effect and an increase in the mitotic fraction. BI 6727 alone reduced clonogenic cell survival, while radiosensitivity in vitro (SF2) and in vivo (single-dose TCD(50) under clamped hypoxia) was not affected. In contrast, local tumour control was significantly improved after application of BI 6727 simultaneously to fractionated irradiation (A431: TCD(50) = 60.5 Gy [95% C.I. 57; 63] after IR alone and <30 Gy after combined treatment; FaDu: 49.5 Gy [43; 56 Gy] versus 32.9 Gy [26; 40]). CONCLUSIONS Despite the lack of direct cellular radiosensitisation, PLK1 inhibition with BI 6727 during fractionated irradiation significantly improves local tumour control when compared to irradiation alone. This result is likely explained by a considerable effect on cell cycle and an independent cytotoxic potential of BI 6727.
Collapse
Affiliation(s)
- Mechthild Krause
- Department of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay-National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), Dresden, Heidelberg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Helmholtz-Zentrum Dresden - Rossendorf.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Vacchelli E, Vitale I, Tartour E, Eggermont A, Sautès-Fridman C, Galon J, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Anticancer radioimmunotherapy. Oncoimmunology 2013; 2:e25595. [PMID: 24319634 PMCID: PMC3850274 DOI: 10.4161/onci.25595] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 06/28/2013] [Indexed: 12/18/2022] Open
Abstract
Radiotherapy has extensively been employed as a curative or palliative intervention against cancer throughout the last century, with a varying degree of success. For a long time, the antineoplastic activity of X- and γ-rays was entirely ascribed to their capacity of damaging macromolecules, in particular DNA, and hence triggering the (apoptotic) demise of malignant cells. However, accumulating evidence indicates that (at least part of) the clinical potential of radiotherapy stems from cancer cell-extrinsic mechanisms, including the normalization of tumor vasculature as well as short- and long-range bystander effects. Local bystander effects involve either the direct transmission of lethal signals between cells connected by gap junctions or the production of diffusible cytotoxic mediators, including reactive oxygen species, nitric oxide and cytokines. Conversely, long-range bystander effects, also known as out-of-field or abscopal effects, presumably reflect the elicitation of tumor-specific adaptive immune responses. Ionizing rays have indeed been shown to promote the immunogenic demise of malignant cells, a process that relies on the spatiotemporally defined emanation of specific damage-associated molecular patterns (DAMPs). Thus, irradiation reportedly improves the clinical efficacy of other treatment modalities such as surgery (both in neo-adjuvant and adjuvant settings) or chemotherapy. Moreover, at least under some circumstances, radiotherapy may potentiate anticancer immune responses as elicited by various immunotherapeutic agents, including (but presumably not limited to) immunomodulatory monoclonal antibodies, cancer-specific vaccines, dendritic cell-based interventions and Toll-like receptor agonists. Here, we review the rationale of using radiotherapy, alone or combined with immunomodulatory agents, as a means to elicit or boost anticancer immune responses, and present recent clinical trials investigating the therapeutic potential of this approach in cancer patients.
Collapse
Affiliation(s)
- Erika Vacchelli
- Gustave Roussy; Villejuif, France
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U848; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
- National Institute of Health; Rome, Italy
| | - Eric Tartour
- INSERM, U970; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
| | | | - Catherine Sautès-Fridman
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
- Equipe 13, Centre de Recherche des Cordeliers; Paris, France
| | - Jérôme Galon
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 15, Centre de Recherche des Cordeliers; Paris, France
- INSERM, U872; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
| | - Laurence Zitvogel
- Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France
- INSERM, U1015; Villejuif, France
| | - Guido Kroemer
- INSERM, U848; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; Assistance Publique-Hôpitaux de Paris; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
- Metabolomics and Cell Biology Platforms; Institut Gustave Roussy; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Equipe 11 labelisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers; Paris, France
| |
Collapse
|
32
|
Koch U, Höhne K, von Neubeck C, Thames HD, Yaromina A, Dahm-Daphi J, Baumann M, Krause M. Residual γH2AX foci predict local tumour control after radiotherapy. Radiother Oncol 2013; 108:434-9. [DOI: 10.1016/j.radonc.2013.06.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 11/28/2022]
|
33
|
Vignard J, Mirey G, Salles B. Ionizing-radiation induced DNA double-strand breaks: a direct and indirect lighting up. Radiother Oncol 2013; 108:362-9. [PMID: 23849169 DOI: 10.1016/j.radonc.2013.06.013] [Citation(s) in RCA: 192] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 06/10/2013] [Accepted: 06/11/2013] [Indexed: 10/26/2022]
Abstract
The occurrence of DNA double-strand breaks (DSBs) induced by ionizing radiation has been extensively studied by biochemical or cell imaging techniques. Cell imaging development relies on technical advances as well as our knowledge of the cell DNA damage response (DDR) process. The DDR involves a complex network of proteins that initiate and coordinate DNA damage signaling and repair activities. As some DDR proteins assemble at DSBs in an established spatio-temporal pattern, visible nuclear foci are produced. In addition, post-translational modifications are important for the signaling and the recruitment of specific partners at damaged chromatin foci. We briefly review here the most widely used methods to study DSBs. We also discuss the development of indirect methods, using reporter expression or intra-nuclear antibodies, to follow the production of DSBs in real time and in living cells.
Collapse
Affiliation(s)
- Julien Vignard
- INRA, UMR1331, Université de Toulouse, TOXALIM (Research Centre in Food Toxicology), F-31027 Toulouse, France
| | | | | |
Collapse
|
34
|
Clinical perspectives of cancer stem cell research in radiation oncology. Radiother Oncol 2013; 108:388-96. [PMID: 23830466 DOI: 10.1016/j.radonc.2013.06.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 06/06/2013] [Indexed: 02/07/2023]
Abstract
Radiotherapy has a proven potential to eradicate cancer stem cells which is reflected by its curative potential in many cancer types. Considerable progress has been made in identification and biological characterisation of cancer stem cells during the past years. Recent biological findings indicate significant inter- and intratumoural and functional heterogeneity of cancer stem cells and lead to more complex models which have potential implications for radiobiology and radiotherapy. Clinical evidence is emerging that biomarkers of cancer stem cells may be prognostic for the outcome of radiotherapy in some tumour entities. Perspectives of cancer stem cell based research for radiotherapy reviewed here include their radioresistance compared to the mass of non-cancer stem cells which form the bulk of all tumour cells, implications for image- and non-image based predictive bio-assays of the outcome of radiotherapy and a combination of novel systemic treatments with radiotherapy.
Collapse
|
35
|
Farjam R, Tsien CI, Feng FY, Gomez-Hassan D, Hayman JA, Lawrence TS, Cao Y. Physiological imaging-defined, response-driven subvolumes of a tumor. Int J Radiat Oncol Biol Phys 2013; 85:1383-90. [PMID: 23257692 PMCID: PMC3638951 DOI: 10.1016/j.ijrobp.2012.10.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Revised: 10/16/2012] [Accepted: 10/23/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE To develop an image analysis framework to delineate the physiological imaging-defined subvolumes of a tumor in relating to treatment response and outcome. METHODS AND MATERIALS Our proposed approach delineates the subvolumes of a tumor based on its heterogeneous distributions of physiological imaging parameters. The method assigns each voxel a probabilistic membership function belonging to the physiological parameter classes defined in a sample of tumors, and then calculates the related subvolumes in each tumor. We applied our approach to regional cerebral blood volume (rCBV) and Gd-DTPA transfer constant (K(trans)) images of patients who had brain metastases and were treated by whole-brain radiation therapy (WBRT). A total of 45 lesions were included in the analysis. Changes in the rCBV (or K(trans))-defined subvolumes of the tumors from pre-RT to 2 weeks after the start of WBRT (2W) were evaluated for differentiation of responsive, stable, and progressive tumors using the Mann-Whitney U test. Performance of the newly developed metrics for predicting tumor response to WBRT was evaluated by receiver operating characteristic (ROC) curve analysis. RESULTS The percentage decrease in the high-CBV-defined subvolumes of the tumors from pre-RT to 2W was significantly greater in the group of responsive tumors than in the group of stable and progressive tumors (P<.007). The change in the high-CBV-defined subvolumes of the tumors from pre-RT to 2W was a predictor for post-RT response significantly better than change in the gross tumor volume observed during the same time interval (P=.012), suggesting that the physiological change occurs before the volumetric change. Also, K(trans) did not add significant discriminatory information for assessing response with respect to rCBV. CONCLUSION The physiological imaging-defined subvolumes of the tumors delineated by our method could be candidates for boost target, for which further development and evaluation is warranted.
Collapse
Affiliation(s)
- Reza Farjam
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI 48109-2099
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
| | - Christina I. Tsien
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
| | - Felix Y. Feng
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
| | - Diana Gomez-Hassan
- Department of Radiology, University of Michigan, 1500 E. Medical Center Drive, Med Inn Building C478, Ann Arbor, MI 48109-5842
| | - James A. Hayman
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
| | - Theodore S. Lawrence
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
| | - Yue Cao
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Blvd., Ann Arbor, MI 48109-2099
- Department of Radiation Oncology, University of Michigan, 1500 E. Medical Center Drive, SPC 5010, Ann Arbor, MI 48109-5010
- Department of Radiology, University of Michigan, 1500 E. Medical Center Drive, Med Inn Building C478, Ann Arbor, MI 48109-5842
| |
Collapse
|
36
|
Horsman MR, Mortensen LS, Petersen JB, Busk M, Overgaard J. Imaging hypoxia to improve radiotherapy outcome. Nat Rev Clin Oncol 2012; 9:674-87. [DOI: 10.1038/nrclinonc.2012.171] [Citation(s) in RCA: 422] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
37
|
Dikomey E, Dahm-Daphi J, Distel L. Prädiktion von Normal- und Tumorreaktion nach Strahlentherapie. Strahlenther Onkol 2012; 188 Suppl 3:304-7. [DOI: 10.1007/s00066-012-0204-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
38
|
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. Radiotherapy is a mainstay of treatment, either alone for early stage tumors or combined with chemotherapy for late stage tumors. An overall 5-year survival rate of around 50% for HNSCC demonstrates that treatment is often unsuccessful. Prediction of outcome is, therefore, aimed at sparing patients from ineffective and toxic treatments on the one hand, and indicating more successful treatment modalities on the other. Both functional and genetic assays have been developed to predict intrinsic radiosensitivity, hypoxia, and repopulation rate. Few, however, have shown consistent correlations with outcome across multiple studies. Messenger RNA and microRNA profiling show promise for predicting hypoxia, whereas epidermal growth factor receptor expression combined with other measures of tumor differentiation grade shows promise for predicting repopulation rate. Intrinsic radiosensitivity assays have not proven useful to date, although development of repair protein foci assays indicates promise from preclinical studies. Assays for cancer stem cell content have shown promise in several clinical studies. In addition, 2 assays showing robustness as predictors for outcome in HNSCC are human papilloma virus status and epidermal growth factor receptor expression. Neither these nor stem cell assays, however, can as yet reliably indicate alternative and better treatments for poor prognosis patients. It would be of great value to have assays that predict the benefit for an individual from combining new molecularly targeted agents with radiotherapy to increase response, in particular those that exploit tumor mutations to provide tumor specificity. Predictive assays are being developed for detecting defects in repair pathways for single- and double-strand DNA breaks, which should allow selection of drugs targeting the appropriate backup pathway, thus exploiting the concept of synthetic lethality. This is one of the most promising areas for prediction, both currently and in the future.
Collapse
|
39
|
Yaromina A, Krause M, Baumann M. Individualization of cancer treatment from radiotherapy perspective. Mol Oncol 2012; 6:211-21. [PMID: 22381063 DOI: 10.1016/j.molonc.2012.01.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 01/21/2012] [Accepted: 01/23/2012] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy is today used in about 50% of all cancer patients, often in multidisciplinary approaches. With major advance in radiotherapy techniques, increasing knowledge on tumor genetics and biology and the continuous introduction of specifically targeted drugs into combined radio-oncological treatment schedules, individualization of radiotherapy is of high priority to further improve treatment outcomes, i.e. to increase long-term tumor cure and/or to reduce chronic treatment toxicity. This review gives an overview on the importance of predictive biomarkers for the field of radiation oncology. The current status of knowledge on potential biomarkers of tumor hypoxia, tumor cell metabolism, DNA repair, cancer stem cells and biomarkers for combining radiotherapy with inhibition of the epidermal growth factor receptor using monoclonal antibodies is described.
Collapse
Affiliation(s)
- Ala Yaromina
- Department of Radiation Oncology, OncoRay-National Center for Radiation Research in Oncology, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | | | | |
Collapse
|