1
|
Kafkaletos A, Sachpazidis I, Mix M, Carles M, Schäfer H, Rühle A, Nicolay NH, Lazzeroni M, Toma-Dasu I, Grosu AL, Baltas D. Implications of the partial volume effect correction on the spatial quantification of hypoxia based on [ 18F]FMISO PET/CT data. Phys Med 2024; 128:104853. [PMID: 39522364 DOI: 10.1016/j.ejmp.2024.104853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
PURPOSE This study evaluates the impact of partial volume effect (PVE) correction on [18F]fluoromisonidazole (FMISO) PET images, focusing on the conversion of standardized uptake values (SUV) to partial oxygen pressure (pO2) and the subsequent determination of hypoxic tumor volume (HTV). METHODS FMISO PET images from 49 head and neck squamous cell carcinoma cases were retrospectively corrected for PVE and converted to pO2. A pO2 threshold of 10 mmHg was used to delineate the HTV (HTVpO2). Comparisons of pO2 distribution and HTVpO2 between corrected and uncorrected images were made, with pO2 distributions evaluated against published polarographic data. HTVpO2 was compared to HTV defined by the conventional tumor-to-muscle ratio (TMR) method (HTVTMR) in terms of volume and topography (DICE coefficient, Hausdorff distance, and center-of-gravity distance) across different TMR cutoff levels. The cutoff level where the segmentation results from both methods were most similar was identified (TMRbest). RESULTS The PVE correction led to decreased minimum pO2, increased HTVpO2 and the identification of more hypoxic cases (HTV > 0). The pO2 distribution demonstrated improved alignment with published polarographic data. At TMRbest 1.6, the center-of-gravity distance between HTVTMR and HTVpO2 demonstrated a low median at 1.5 mm, while the wide range (0.0 to 9.6 mm) indicated high interpatient variability. The shape of HTV exhibited considerable variation with DICE 0.74 (0.03 to 1.00) and Hausdorff distance 8.5 mm (2.0 to 42.8 mm). CONCLUSIONS PVE correction is recommended before converting SUV to pO2 for the spatially resolved quantification of hypoxia.
Collapse
Affiliation(s)
- Athanasios Kafkaletos
- Division of Medical Physics, Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany.
| | - Ilias Sachpazidis
- Division of Medical Physics, Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany
| | - Montserrat Carles
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), Valencia, Spain
| | - Henning Schäfer
- Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany; Department of Radiation Oncology, University of Leipzig Medical Centre, Leipzig, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, University of Leipzig Medical Centre, Leipzig, Germany
| | - Marta Lazzeroni
- Physics Department, Stockholm University, Sweden; Oncology-Pathology Department, Karolinska Institute, Stockholm, Sweden
| | - Iuliana Toma-Dasu
- Physics Department, Stockholm University, Sweden; Oncology-Pathology Department, Karolinska Institute, Stockholm, Sweden
| | - Anca L Grosu
- Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany
| | - Dimos Baltas
- Division of Medical Physics, Department of Radiation Oncology, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, Germany
| |
Collapse
|
2
|
Jiang X, Xu XN, Yuan XY, Jiang HR, Zhao MJ, Duan YX, Li G. The apparent diffusion coefficient can serve as a predictor of survival in patients with gliomas. Radiat Oncol 2024; 19:149. [PMID: 39472956 PMCID: PMC11524024 DOI: 10.1186/s13014-024-02535-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND AND PURPOSE Magnetic resonance imaging is indispensable for the preoperative diagnosis of glioma. This study aimed to investigate the role of the apparent diffusion coefficient values as predictors of survival in patients with gliomas. METHODS AND MATERIALS A retrospective analysis was conducted on 101 patients with gliomas who underwent surgery between 2015 and 2020. Diffusion-weighted MRI was performed before the surgery. The regions of interest were categorized into parenchymal area, non-enhancing peritumoral area, and necrotic or cystic area. All the patients were divided into three subgroups: the parenchyma group, the non-enhancing peritumoral signal abnormality group, and the necrosis or cyst group. Univariate and multivariate analyses were performed using COX regression. RESULTS In the parenchymal group, Ki67, P53, IDH, and the high or low ADC values were identified as independent prognosticators for disease-free survival, while Ki67, IDH, and the high or low ADC values for overall survival. In the non-enhancing peritumoral signal abnormality group, Ki67, P53, IDH, and the ADC parenchymal area/ADC non-enhancing peritumoral area ratio were identified as independent prognostic factors for disease-free survival, while Ki67, IDH, and the ADC parenchymal area/ADC non-enhancing peritumoral area ratio for overall survival. In the necrosis or cyst group, Ki67 was significantly associated with disease-free survival, while Ki67 and the ADC value of the necrotic or cystic area for overall survival. CONCLUSIONS The ADC values, including the ADC value in the parenchymal area, the ADC parenchymal area/ADC non-enhancing peritumoral area ratio, and the ADC value in the necrotic or cystic area, can serve as an efficient and potential index for predicting the survival of patients with glioma.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Pathology, Jinhua Municipal Central Hospital, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xu-Ni Xu
- Department of Radiology, Shaoxing Central Hospital, The Central Hospital of Shaoxing University, Shaoxing, Zhejiang, 312030, China
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xiao-Ye Yuan
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Hao-Ran Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Meng-Jing Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yu-Xia Duan
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Gang Li
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
3
|
De Bruycker A, De Neve W, Daisne JF, Vercauteren T, De Gersem W, Olteanu L, Berwouts D, Deheneffe S, Madani I, Goethals I, Duprez F. Disease Control and Late Toxicity in Adaptive Dose Painting by Numbers Versus Nonadaptive Radiation Therapy for Head and Neck Cancer: A Randomized Controlled Phase 2 Trial. Int J Radiat Oncol Biol Phys 2024; 120:516-527. [PMID: 38387811 DOI: 10.1016/j.ijrobp.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2023] [Accepted: 01/02/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Local recurrence remains the main cause of death in stage III-IV nonmetastatic head and neck cancer (HNC), with relapse-prone regions within high 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET)-signal gross tumor volume. We investigated if dose escalation within this subvolume combined with a 3-phase treatment adaptation could increase local (LC) and regional (RC) control at equal or minimized radiation-induced toxicity, by comparing adaptive 18F-FDG-PET voxel intensity-based dose painting by numbers (A-DPBN) with nonadaptive standard intensity modulated radiation therapy (S-IMRT). METHODS AND MATERIALS This 2-center randomized controlled phase 2 trial assigned (1:1) patients to receive A-DPBN or S-IMRT (+/-chemotherapy). Eligibility: nonmetastatic HNC of oral cavity, oro-/hypopharynx, or larynx, needing radio(chemo)therapy; T1-4N0-3 (exception: T1-2N0 glottic); KPS ≥ 70; ≥18 years; and informed consent. PRIMARY OUTCOMES 1-year LC and RC. The dose prescription for A-DPBN was intercurrently adapted in 2 steps to an absolute dose-volume limit (≤1.75 cm3 can receive >84 Gy and normalized isoeffective dose >96 Gy) as a safety measure during the study course after 4/7 A-DPBN patients developed ≥G3 mucosal ulcers. RESULTS Ninety-five patients were randomized (A-DPBN, 47; S-IMRT, 48). Median follow-up was 31 months (IQR, 14-48 months); 29 patients died (17 of cancer progression). A-DPBN resulted in superior LC compared with S-IMRT, with 1- and 2-year LC of 91% and 88% versus 78% and 75%, respectively (hazard ratio, 3.13; 95% CI, 1.13-8.71; P = .021). RC and overall survival were comparable between arms, as was overall grade (G) ≥3 late toxicity (36% vs 20%; P = .1). More ≥G3 late mucosal ulcers were observed in active smokers (29% vs 3%; P = .005) and alcohol users (33% vs 13%; P = .02), independent of treatment arm. Similarly, in the A-DPBN arm, significantly more patients who smoked at diagnosis developed ≥G3 (46% vs 12%; P = .005) and ≥G4 (29% vs 8%; P = .048) mucosal ulcers. One arterial blowout occurred after a G5 mucosal toxicity. CONCLUSIONS A-DPBN resulted in superior 1- and 2-year LC for HNC compared with S-IMRT. This supports further exploration in multicenter phase 3 trials. It will, however, be challenging to recruit a substantial patient sample for such trials, as concerns have arisen regarding the association of late mucosal ulcers when escalating the dose in continuing smokers.
Collapse
Affiliation(s)
- Aurélie De Bruycker
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium.
| | - Wilfried De Neve
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Jean-François Daisne
- Department of Radiation Oncology, Université Catholique de Louvain, CHU-UCL-Namur, Namur, Belgium; Department of Radiation Oncology, University Hospital Leuven, Leuven, Belgium; Department of Oncology, Leuven Cancer Institute (LKI), Catholic University of Leuven, Leuven, Belgium
| | - Tom Vercauteren
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Werner De Gersem
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Luiza Olteanu
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Dieter Berwouts
- Department of Nuclear Medicine, AZ Maria-Middelares, AZ Jan Palfijn, Ghent, Belgium
| | - Stéphanie Deheneffe
- Department of Radiation Oncology, Université Catholique de Louvain, CHU-UCL-Namur, Namur, Belgium
| | - Indira Madani
- Department of Radiation Oncology, University Hospital of Zurich, Zurich, Switzerland
| | - Ingeborg Goethals
- Faculty of Medicine and Health Sciences, Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Fréderic Duprez
- Department of Radiation Oncology, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
4
|
Sambasivan K, Barrington SF, Connor SE, Witney TH, Blower PJ, Urbano TG. Is there a role for [ 18F]-FMISO PET to guide dose adaptive radiotherapy in head and neck cancer? A review of the literature. Clin Transl Imaging 2024; 12:137-155. [PMID: 39286295 PMCID: PMC7616449 DOI: 10.1007/s40336-023-00607-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/12/2023] [Indexed: 09/19/2024]
Abstract
Purpose Hypoxia is a major cause of radioresistance in head and neck cancer (HNC), resulting in treatment failure and disease recurrence. 18F-fluoromisonidazole [18F]FMISO PET has been proposed as a means of localising intratumoural hypoxia in HNC so that radiotherapy can be specifically escalated in hypoxic regions. This concept may not be deliverable in routine clinical practice, however, given that [18F]FMISO PET is costly, time consuming and difficult to access. The aim of this review was to summarise clinical studies involving [18F]FMISO PET to ascertain whether it can be used to guide radiotherapy treatment in HNC. Methods A comprehensive literature search was conducted on PubMed and Web of Science databases. Studies investigating [18F]FMISO PET in newly diagnosed HNC patients were considered eligible for review. Results We found the following important results from our literature review: 1)Studies have focussed on comparing [18F]FMISO PET to other hypoxia biomarkers, but currently there is no evidence of a strong correlation between [18F]FMISO and these biomarkers.2)The results of [18F]FMISO PET imaging are not necessarily repeatable, and the location of uptake may vary during treatment.3)Tumour recurrences do not always occur within the pretreatment hypoxic volume on [18F]FMISO PET.4)Dose modification studies using [18F]FMISO PET are in a pilot phase and so far, none have demonstrated the efficacy of radiotherapy dose painting according to [18F]FMISO uptake on PET. Conclusions Our results suggest it is unlikely [18F]FMISO PET will be suitable for radiotherapy dose adaptation in HNC in a routine clinical setting. Part of the problem is that hypoxia is a dynamic phenomenon, and thus difficult to delineate on a single scan. Currently, it is anticipated that [18F]FMISO PET will remain useful within the research setting only.
Collapse
Affiliation(s)
- Khrishanthne Sambasivan
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre; School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - Steve Ej Connor
- Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, UK Department of Radiology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London, UK
| | - Timothy H Witney
- King's College London, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, London, United Kingdom
| | - Philip J Blower
- King's College London, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, London, United Kingdom
| | - Teresa Guerrero Urbano
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK; Faculty of Dentistry, Oral & Craniofacial Sciences and School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
5
|
McDonald BA, Dal Bello R, Fuller CD, Balermpas P. The Use of MR-Guided Radiation Therapy for Head and Neck Cancer and Recommended Reporting Guidance. Semin Radiat Oncol 2024; 34:69-83. [PMID: 38105096 PMCID: PMC11372437 DOI: 10.1016/j.semradonc.2023.10.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Although magnetic resonance imaging (MRI) has become standard diagnostic workup for head and neck malignancies and is currently recommended by most radiological societies for pharyngeal and oral carcinomas, its utilization in radiotherapy has been heterogeneous during the last decades. However, few would argue that implementing MRI for annotation of target volumes and organs at risk provides several advantages, so that implementation of the modality for this purpose is widely accepted. Today, the term MR-guidance has received a much broader meaning, including MRI for adaptive treatments, MR-gating and tracking during radiotherapy application, MR-features as biomarkers and finally MR-only workflows. First studies on treatment of head and neck cancer on commercially available dedicated hybrid-platforms (MR-linacs), with distinct common features but also differences amongst them, have also been recently reported, as well as "biological adaptation" based on evaluation of early treatment response via functional MRI-sequences such as diffusion weighted ones. Yet, all of these approaches towards head and neck treatment remain at their infancy, especially when compared to other radiotherapy indications. Moreover, the lack of standardization for reporting MR-guided radiotherapy is a major obstacle both to further progress in the field and to conduct and compare clinical trials. Goals of this article is to present and explain all different aspects of MR-guidance for radiotherapy of head and neck cancer, summarize evidence, as well as possible advantages and challenges of the method and finally provide a comprehensive reporting guidance for use in clinical routine and trials.
Collapse
Affiliation(s)
- Brigid A McDonald
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Riccardo Dal Bello
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Clifton D Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Zurich and University of Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Almhagen E, Dasu A, Johansson S, Traneus E, Ahnesjö A. Plan robustness and RBE influence for proton dose painting by numbers for head and neck cancers. Phys Med 2023; 115:103157. [PMID: 37939480 DOI: 10.1016/j.ejmp.2023.103157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 08/25/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
PURPOSE To investigate the feasibility of dose painting by numbers (DPBN) with respect to robustness for proton therapy for head and neck cancers (HNC), and to study the influence of variable RBE on the TCP and OAR dose burden. METHODS AND MATERIALS Data for 19 patients who have been scanned pretreatment with PET-FDG and subsequently treated with photon therapy were used in the study. A dose response model developed for photon therapy was implemented in a TPS, allowing DPBN plans to be created. Conventional homogeneous dose and DPBN plans were created for each patient, optimized with either fixed RBE = 1.1 or a variable RBE model. Robust optimization was used to create clinically acceptable plans. To estimate the maximum potential loss in TCP due to actual SUV variations from the pre-treatment imaging, we applied a test case with randomized SUV distribution. RESULTS Regardless of the use of variable RBE for optimization or evaluation, a statistically significant increase (p < 0.001) in TCP was found for DPBN plans as compared to homogeneous dose plans. Randomizing the SUV distribution decreased the TCP for all plans. A correlation between TCP increase and variance of the SUV distribution and target volume was also found. CONCLUSION DPBN for protons and HNC is feasible and could lead to a TCP gain. Risks associated with the temporal variation of SUV distributions could be mitigated by imposing minimum doses to targets. The correlation found between TCP increase and SUV variance and target volume may be used for patient selection.
Collapse
Affiliation(s)
- Erik Almhagen
- Medical Radiation Sciences, Department of Immunology, Genetics and Pathology, Uppsala University, Akademiska Sjukhuset, Uppsala, Sweden; The Skandion Clinic, Uppsala, Sweden.
| | - Alexandru Dasu
- Medical Radiation Sciences, Department of Immunology, Genetics and Pathology, Uppsala University, Akademiska Sjukhuset, Uppsala, Sweden; The Skandion Clinic, Uppsala, Sweden
| | - Silvia Johansson
- Divison of Oncology, Department of Immunology, Genetics and Pathology, Uppsala University Hospital, Uppsala, Sweden
| | | | - Anders Ahnesjö
- Medical Radiation Sciences, Department of Immunology, Genetics and Pathology, Uppsala University, Akademiska Sjukhuset, Uppsala, Sweden
| |
Collapse
|
7
|
Rosen BS, Vaishampayan N, Cao Y, Mierzwa ML. The Utility of Interim Positron Emission Tomography Imaging to Inform Adaptive Radiotherapy for Head and Neck Squamous Cell Carcinoma. Cancer J 2023; 29:243-247. [PMID: 37471616 DOI: 10.1097/ppo.0000000000000669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023]
Abstract
ABSTRACT In this article, as part of this special issue on biomarkers of early response, we review the current evidence to support the use of positron emission tomography (PET) imaging during chemoradiation therapy to inform biologically adaptive radiotherapy for head and neck squamous cell carcinoma. We review literature covering this topic spanning nearly 3 decades, including the use of various radiotracers and discoveries of novel predictive PET biomarkers. Through understanding how observational trials have informed current interventional clinical trials, we hope that this review will encourage researchers and clinicians to incorporate PET response criteria in new trial designs to advance biologically optimized radiotherapy.
Collapse
Affiliation(s)
- Benjamin S Rosen
- From the Department of Radiation Oncology, University of Michigan, Ann Arbor, MI
| | | | | | | |
Collapse
|
8
|
Abstract
Hypoxia (oxygen deprivation) occurs in most solid malignancies, albeit with considerable heterogeneity. Hypoxia is associated with an aggressive cancer phenotype by promotion of genomic instability, evasion of anti-cancer therapies including radiotherapy and enhancement of metastatic risk. Therefore, hypoxia results in poor cancer outcomes. Targeting hypoxia to improve cancer outcomes is an attractive therapeutic strategy. Hypoxia-targeted dose painting escalates radiotherapy dose to hypoxic sub-volumes, as quantified and spatially mapped using hypoxia imaging. This therapeutic approach could overcome hypoxia-induced radioresistance and improve patient outcomes without the need for hypoxia-targeted drugs. This article will review the premise and underpinning evidence for personalized hypoxia-targeted dose painting. It will present data on relevant hypoxia imaging biomarkers, highlight the challenges and potential benefit of this approach and provide recommendations for future research priorities in this field. Personalized hypoxia-based radiotherapy de-escalation strategies will also be addressed.
Collapse
Affiliation(s)
- Ahmed Salem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, Hashemite University, Zarqa, Jordan; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
9
|
Schwenck J, Sonanini D, Cotton JM, Rammensee HG, la Fougère C, Zender L, Pichler BJ. Advances in PET imaging of cancer. Nat Rev Cancer 2023:10.1038/s41568-023-00576-4. [PMID: 37258875 DOI: 10.1038/s41568-023-00576-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 06/02/2023]
Abstract
Molecular imaging has experienced enormous advancements in the areas of imaging technology, imaging probe and contrast development, and data quality, as well as machine learning-based data analysis. Positron emission tomography (PET) and its combination with computed tomography (CT) or magnetic resonance imaging (MRI) as a multimodality PET-CT or PET-MRI system offer a wealth of molecular, functional and morphological data with a single patient scan. Despite the recent technical advances and the availability of dozens of disease-specific contrast and imaging probes, only a few parameters, such as tumour size or the mean tracer uptake, are used for the evaluation of images in clinical practice. Multiparametric in vivo imaging data not only are highly quantitative but also can provide invaluable information about pathophysiology, receptor expression, metabolism, or morphological and functional features of tumours, such as pH, oxygenation or tissue density, as well as pharmacodynamic properties of drugs, to measure drug response with a contrast agent. It can further quantitatively map and spatially resolve the intertumoural and intratumoural heterogeneity, providing insights into tumour vulnerabilities for target-specific therapeutic interventions. Failure to exploit and integrate the full potential of such powerful imaging data may lead to a lost opportunity in which patients do not receive the best possible care. With the desire to implement personalized medicine in the cancer clinic, the full comprehensive diagnostic power of multiplexed imaging should be utilized.
Collapse
Affiliation(s)
- Johannes Schwenck
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Dominik Sonanini
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Jonathan M Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Department of Immunology, IFIZ Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany
- Medical Oncology and Pulmonology, Department of Internal Medicine, Eberhard Karls University of Tübingen, Tübingen, Germany
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University of Tübingen, Tübingen, Germany.
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided and Functionally Instructed Tumour Therapies', Eberhard Karls University, Tübingen, Germany.
- German Cancer Research Center, German Cancer Consortium DKTK, Partner Site Tübingen, Tübingen, Germany.
| |
Collapse
|
10
|
Hildingsson S, Gebre-Medhin M, Zschaeck S, Adrian G. Hypoxia in relationship to tumor volume using hypoxia PET-imaging in head & neck cancer - A scoping review. Clin Transl Radiat Oncol 2022; 36:40-46. [PMID: 35769424 PMCID: PMC9234341 DOI: 10.1016/j.ctro.2022.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023] Open
Abstract
Primary tumor volume and hypoxic volume has previously not been convincingly related. 367 patients with head and neck squamous cell carcinoma from 21 different studies using hypoxia-PET The hypoxic volume increased significantly with primary tumor volume. In larger tumor the hypoxic fraction was significantly higher than in smaller tumors.
Background Hypoxia and large tumor volumes are negative prognostic factors for patients with head and neck squamous cell carcinoma (HNSCC) treated with radiation therapy (RT). PET-scanning with specific hypoxia-tracers (hypoxia-PET) can be used to non-invasively assess hypoxic tumor volume. Primary tumor volume is readily available for patients undergoing RT. However, the relationship between hypoxic volume and primary tumor volume is yet an open question. The current study investigates the hypotheses that larger tumors contain both a larger hypoxic volume and a higher hypoxic fraction. Methods PubMed and Embase were systematically searched to identify articles fulfilling the predefined criteria. Individual tumor data (primary tumor volume and hypoxic volume/fraction) was extracted. Relationship between hypoxic volume and primary tumor volume was investigated by linear regression. The correlation between hypoxic fraction and log2(primary tumor volume) was determined for each cohort and in a pooled analysis individual regression slopes and coefficients of determination (R2) were weighted according to cohort size. Results 21 relevant articles were identified and individual data from 367 patients was extracted, out of which 323 patients from 17 studies had quantifiable volumes of interest. A correlation between primary tumor volume and PET-determined hypoxic volume was found (P <.001, R2 = 0.46). Larger tumors had a significantly higher fraction of hypoxia compared with smaller tumors (P<.01). The weighted analysis of all studies revealed that for each doubling of the tumor volume, the hypoxic fraction increased by four percentage points. Conclusion This study shows correlations between primary tumor volume and hypoxic volume as well as primary tumor volume and the hypoxic fraction in patients with HNSCC. The findings suggest that not only do large tumors contain more cancer cells, they also have a higher proportion of potentially radioresistant hypoxic cells. This knowledge can be important when individualizing RT.
Collapse
Affiliation(s)
- Sofia Hildingsson
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Gebre-Medhin
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gabriel Adrian
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
11
|
Welz S, Paulsen F, Pfannenberg C, Reimold M, Reischl G, Nikolaou K, La Fougère C, Alber M, Belka C, Zips D, Thorwarth D. Dose escalation to hypoxic subvolumes in head and neck cancer: A randomized phase II study using dynamic [ 18F]FMISO PET/CT. Radiother Oncol 2022; 171:30-36. [PMID: 35395276 DOI: 10.1016/j.radonc.2022.03.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Tumor hypoxia is a major cause of resistance to radiochemotherapy in locally advanced head-and-neck cancer (LASCCHN). We present results of a randomized phase II trial on hypoxia dose escalation (DE) in LASCCHN based on dynamic [18F]FMISO (dynFMISO) positron emission tomography (PET). The purpose was to confirm the prognostic value of hypoxia PET and assess feasibility, toxicity and efficacy of hypoxia-DE. MATERIALS AND METHODS Patients with LASCCHN underwent baseline dynFMISO PET/CT. Hypoxic volumes (HV) were derived from dynFMISO data. Patients with hypoxic tumors (HV>0) were randomized into standard radiotherapy (ST: 70Gy/35fx) or dose escalation (DE: 77Gy/35fx) to the HV. Patients with non-hypoxic tumors were treated with ST. After a minimum follow-up of 2 years, feasibility, acute/late toxicity and local control (LC) were analyzed. RESULTS The study was closed prematurely due to slow accrual. Between 2009 and 2017, 53 patients were enrolled, 39 (74%) had hypoxic tumors and were randomized into ST or DE. For non-hypoxic patients, 100% 5-year LC was observed compared to 74% in patients with hypoxic tumors (p=0.039). The difference in 5-year LC between DE (16/19) and ST (10/17) was 25%, p=0.150. No relevant differences related to acute and late toxicities between the groups were observed. CONCLUSION This study confirmed the prognostic value of hypoxia PET in LASCCHN for LC. Outcome after hypoxia DE appears promising and may support the concept of DE. Slow accrual and premature closure may partly be due to a high complexity of the study setup which needs to be considered for future multicenter trials.
Collapse
Affiliation(s)
- Stefan Welz
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Matthias Reimold
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christian La Fougère
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Alber
- Section for Medical Physics, Department of Radiation Oncology, Heidelberg University, Heidelberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University of Munich, Germany; Department of Radiation Oncology, LMU Munich, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Wright P, Arnesen MR, Lønne PI, Suilamo S, Silvoniemi A, Dale E, Minn H, Malinen E. Repeatability of hypoxia dose painting by numbers based on EF5-PET in head and neck cancer. Acta Oncol 2021; 60:1386-1391. [PMID: 34184605 DOI: 10.1080/0284186x.2021.1944663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Hypoxia dose painting is a radiotherapy technique to increase the dose to hypoxic regions of the tumour. Still, the clinical effect relies on the reproducibility of the hypoxic region shown in the medical image. 18F-EF5 is a hypoxia tracer for positron emission tomography (PET), and this study investigated the repeatability of 18F-EF5-based dose painting by numbers (DPBN) in head and neck cancer (HNC). MATERIALS AND METHODS Eight HNC patients undergoing two 18F-EF5-PET/CT sessions (A and B) before radiotherapy were included. A linear conversion of PET signal intensity to radiotherapy dose prescription was employed and DPBN treatment plans were created using the image basis acquired at each PET/CT session. Also, plan A was recalculated on the image basis for session B. Voxel-by-voxel Pearson's correlation and quality factor were calculated to assess the DPBN plan quality and repeatability. RESULTS The mean (SD) correlation coefficient between DPBN prescription and plan was 0.92 (0.02) and 0.93 (0.02) for sessions A and B, respectively, with corresponding quality factors of 0.02 (0.002) and 0.02 (0.003), respectively. The mean correlation between dose prescriptions at day A and B was 0.72 (0.13), and 0.77 (0.12) for the corresponding plans. A mean correlation of 0.80 (0.08) was found between plan A, recalculated on image basis B, and plan B. CONCLUSION Hypoxia DPBN planning based on 18F-EF5-PET/CT showed high repeatability. This illustrates that 18F-EF5-PET provides a robust target for dose painting.
Collapse
Affiliation(s)
- Pauliina Wright
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | | | - Per-Ivar Lønne
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Sami Suilamo
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Antti Silvoniemi
- Department of Otorhinolaryngology-Head and Neck Surgery, Turku University Hospital, Turku PET Centre, University of Turku, Turku, Finland
| | - Einar Dale
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Heikki Minn
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku PET Centre, University of Turku, Turku, Finland
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Incidental uptake of fluorodeoxyglucose in the Waldeyer's ring and risk of oropharyngeal malignancy. Eur Arch Otorhinolaryngol 2021; 279:2657-2664. [PMID: 34570264 PMCID: PMC8986689 DOI: 10.1007/s00405-021-07089-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/12/2021] [Indexed: 10/27/2022]
Abstract
PURPOSE Fluorodeoxyglucose (FDG) positron emission tomography (PET) is increasingly used to diagnose and stage malignancy. The aim of this article is to investigate the significance of incidental FDG uptake in the Waldeyer's ring and to assess its value in predicting clinically occult oropharyngeal malignancy. METHODS All FDG-PET/CT scans performed in Imperial College NHS Foundation Trust, UK between January 2012 and November 2018 were included. Patients with known or suspected oropharyngeal malignancy or lymphoma were excluded. Minimum follow-up was 12 months. RESULTS A total of 724 scans revealed oropharyngeal uptake of FDG. Of these, 102 were included in the study. Most patients (62.1%) were scanned as part of staging for other malignancies. Oropharyngeal FDG uptake was asymmetrical in 57.3% of the cases. Uptake was more common in the tonsils (56.3%), followed by the tongue base (31.1%) and both sites (12.6%). In 41.7% of reports, appearance was described as likely physiological; however, 52.4% of reports advised direct visualisation, clinical correlation or ENT opinion. Only 24.3% (25/102) of patients were referred and seen by ENT, 14.6% (15/102) of which had an interval PET scan and 8.7% (9/102) proceeded to tissue diagnosis. There was one oropharyngeal cancer identified and one unexpected metastasis from esophageal cancer. CONCLUSION Incidental uptake on PET/CT in the oropharynx is common. However, malignancy is rare (1.9%) and, when present, is associated with high SUVmax and asymmetrical uptake. Imaging results must be correlated clinically. These patients should be seen by an ENT specialist yet most may not require further investigations.
Collapse
|
14
|
Elamir AM, Stanescu T, Shessel A, Tadic T, Yeung I, Letourneau D, Kim J, Lukovic J, Dawson LA, Wong R, Barry A, Brierley J, Gallinger S, Knox J, O'Kane G, Dhani N, Hosni A, Taylor E. Simulated dose painting of hypoxic sub-volumes in pancreatic cancer stereotactic body radiotherapy. Phys Med Biol 2021; 66. [PMID: 34438383 DOI: 10.1088/1361-6560/ac215c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/26/2021] [Indexed: 12/26/2022]
Abstract
Dose painting of hypoxic tumour sub-volumes using positron-emission tomography (PET) has been shown to improve tumour controlin silicoin several sites, predominantly head and neck and lung cancers. Pancreatic cancer presents a more stringent challenge, given its proximity to critical gastro-intestinal organs-at-risk (OARs), anatomic motion, and impediments to reliable PET hypoxia quantification. A radiobiological model was developed to estimate clonogen survival fraction (SF), using18F-fluoroazomycin arabinoside PET (FAZA PET) images from ten patients with unresectable pancreatic ductal adenocarcinoma to quantify oxygen enhancement effects. For each patient, four simulated five-fraction stereotactic body radiotherapy (SBRT) plans were generated: (1) a standard SBRT plan aiming to cover the planning target volume with 40 Gy, (2) dose painting plans delivering escalated doses to a maximum of three FAZA-avid hypoxic sub-volumes, (3) dose painting plans with simulated spacer separating the duodenum and pancreatic head, and (4), plans with integrated boosts to geometric contractions of the gross tumour volume (GTV). All plans saturated at least one OAR dose limit. SF was calculated for each plan and sensitivity of SF to simulated hypoxia quantification errors was evaluated. Dose painting resulted in a 55% reduction in SF as compared to standard SBRT; 78% with spacer. Integrated boosts to hypoxia-blind geometric contractions resulted in a 41% reduction in SF. The reduction in SF for dose-painting plans persisted for all hypoxia quantification parameters studied, including registration and rigid motion errors that resulted in shifts and rotations of the GTV and hypoxic sub-volumes by as much as 1 cm and 10 degrees. Although proximity to OARs ultimately limited dose escalation, with estimated SFs (∼10-5) well above levels required to completely ablate a ∼10 cm3tumour, dose painting robustly reduced clonogen survival when accounting for expected treatment and imaging uncertainties and thus, may improve local response and associated morbidity.
Collapse
Affiliation(s)
- Ahmed M Elamir
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Teodor Stanescu
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Andrea Shessel
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada
| | - Tony Tadic
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Ivan Yeung
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada.,Stronach Regional Cancer Centre, Southlake Regional Health Centre, Newmarket, Canada
| | - Daniel Letourneau
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - John Kim
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Jelena Lukovic
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Laura A Dawson
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Rebecca Wong
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Aisling Barry
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - James Brierley
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Steven Gallinger
- Ontario Institute for Cancer Research, PanCuRx Translational Research Initiative, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada
| | - Jennifer Knox
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Grainne O'Kane
- Ontario Institute for Cancer Research, PanCuRx Translational Research Initiative, Toronto, Canada.,Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Neesha Dhani
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Center, Toronto, Canada.,Department of Medicine, University of Toronto, Toronto, Canada
| | - Ali Hosni
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - Edward Taylor
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, Canada
| |
Collapse
|
15
|
Wang C, Padgett KR, Su MY, Mellon EA, Maziero D, Chang Z. Multi-parametric MRI (mpMRI) for treatment response assessment of radiation therapy. Med Phys 2021; 49:2794-2819. [PMID: 34374098 DOI: 10.1002/mp.15130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 11/11/2022] Open
Abstract
Magnetic resonance imaging (MRI) plays an important role in the modern radiation therapy (RT) workflow. In comparison with computed tomography (CT) imaging, which is the dominant imaging modality in RT, MRI possesses excellent soft-tissue contrast for radiographic evaluation. Based on quantitative models, MRI can be used to assess tissue functional and physiological information. With the developments of scanner design, acquisition strategy, advanced data analysis, and modeling, multiparametric MRI (mpMRI), a combination of morphologic and functional imaging modalities, has been increasingly adopted for disease detection, localization, and characterization. Integration of mpMRI techniques into RT enriches the opportunities to individualize RT. In particular, RT response assessment using mpMRI allows for accurate characterization of both tissue anatomical and biochemical changes to support decision-making in monotherapy of radiation treatment and/or systematic cancer management. In recent years, accumulating evidence have, indeed, demonstrated the potentials of mpMRI in RT response assessment regarding patient stratification, trial benchmarking, early treatment intervention, and outcome modeling. Clinical application of mpMRI for treatment response assessment in routine radiation oncology workflow, however, is more complex than implementing an additional imaging protocol; mpMRI requires additional focus on optimal study design, practice standardization, and unified statistical reporting strategy to realize its full potential in the context of RT. In this article, the mpMRI theories, including image mechanism, protocol design, and data analysis, will be reviewed with a focus on the radiation oncology field. Representative works will be discussed to demonstrate how mpMRI can be used for RT response assessment. Additionally, issues and limits of current works, as well as challenges and potential future research directions, will also be discussed.
Collapse
Affiliation(s)
- Chunhao Wang
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| | - Kyle R Padgett
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA.,Department of Radiology, University of Miami, Miami, Florida, USA
| | - Min-Ying Su
- Department of Radiological Sciences, University of California, Irvine, California, USA.,Department of Medical Imaging and Radiological Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Eric A Mellon
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Danilo Maziero
- Department of Radiation Oncology, University of Miami, Miami, Florida, USA
| | - Zheng Chang
- Department of Radiation Oncology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
16
|
Carles M, Fechter T, Grosu AL, Sörensen A, Thomann B, Stoian RG, Wiedenmann N, Rühle A, Zamboglou C, Ruf J, Martí-Bonmatí L, Baltas D, Mix M, Nicolay NH. 18F-FMISO-PET Hypoxia Monitoring for Head-and-Neck Cancer Patients: Radiomics Analyses Predict the Outcome of Chemo-Radiotherapy. Cancers (Basel) 2021; 13:3449. [PMID: 34298663 PMCID: PMC8303992 DOI: 10.3390/cancers13143449] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia is associated with radiation resistance and can be longitudinally monitored by 18F-fluoromisonidazole (18F-FMISO)-PET/CT. Our study aimed at evaluating radiomics dynamics of 18F-FMISO-hypoxia imaging during chemo-radiotherapy (CRT) as predictors for treatment outcome in head-and-neck squamous cell carcinoma (HNSCC) patients. We prospectively recruited 35 HNSCC patients undergoing definitive CRT and longitudinal 18F-FMISO-PET/CT scans at weeks 0, 2 and 5 (W0/W2/W5). Patients were classified based on peritherapeutic variations of the hypoxic sub-volume (HSV) size (increasing/stable/decreasing) and location (geographically-static/geographically-dynamic) by a new objective classification parameter (CP) accounting for spatial overlap. Additionally, 130 radiomic features (RF) were extracted from HSV at W0, and their variations during CRT were quantified by relative deviations (∆RF). Prediction of treatment outcome was considered statistically relevant after being corrected for multiple testing and confirmed for the two 18F-FMISO-PET/CT time-points and for a validation cohort. HSV decreased in 64% of patients at W2 and in 80% at W5. CP distinguished earlier disease progression (geographically-dynamic) from later disease progression (geographically-static) in both time-points and cohorts. The texture feature low grey-level zone emphasis predicted local recurrence with AUCW2 = 0.82 and AUCW5 = 0.81 in initial cohort (N = 25) and AUCW2 = 0.79 and AUCW5 = 0.80 in validation cohort. Radiomics analysis of 18F-FMISO-derived hypoxia dynamics was able to predict outcome of HNSCC patients after CRT.
Collapse
Affiliation(s)
- Montserrat Carles
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Tobias Fechter
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Anca L. Grosu
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Arnd Sörensen
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Benedikt Thomann
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Raluca G. Stoian
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nicole Wiedenmann
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexander Rühle
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Constantinos Zamboglou
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Juri Ruf
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Luis Martí-Bonmatí
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Dimos Baltas
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Michael Mix
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Nuclear Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nils H. Nicolay
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
17
|
Kawamura M, Yoshimura M, Shimizu Y, Sano K, Ishimori T, Nakamoto Y, Mizowaki T, Hiraoka M. Evaluation of Optimal Post-Injection Timing of Hypoxic Imaging with 18F-Fluoromisonidazole-PET/CT. Mol Imaging Biol 2021; 23:597-603. [PMID: 33475945 DOI: 10.1007/s11307-021-01580-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/26/2020] [Accepted: 01/06/2021] [Indexed: 11/29/2022]
Abstract
PURPOSE Positron emission tomography (PET)/computed tomography (CT) using 18F-fluoromisonidazole (FMISO) has been used as an imaging tool for tumour hypoxia. However, it remains unclear whether they are useful when scanning is performed earlier, e.g. at 2-h post-injection with a high sensitivity PET scanner. This study aimed to investigate the relationship between quantitative values in 18F-fluoromisonidazole (18F-FMISO)-PET obtained at 2- and 4-h post-injection in patients with head and neck cancer. PROCEDURES We enrolled 20 patients with untreated locally advanced head and neck cancer who underwent 18F-FMISO-PET/CT scan between August 2015 and March 2018 at our institute. Image acquisition was performed 2 h and 4 h after 18F-FMISO administration using a combined PET/CT scanner. The SUVmax, SUVmean, SUVpeak, tumour-to-blood ratio (TBR), tumour-to-muscle ratio (TMR), metabolic tumour volume (MTV), and total lesion hypoxia (TLH) were measured in the region of interest of the primary tumour. We evaluated the between-image Spearman's rank correlation coefficients and percentage differences in the quantitative values. The locations of the maximum uptake pixel were identified in both scans, and the distance between them was measured. RESULTS The mean (SD) SUVmax at 2 h and 4 h was 2.2(0.7) and 2.4(0.8), respectively. The Spearman's rank correlation coefficients (ρ) and mean (SD) of the percentage differences of the measures were as follows: SUVmax (0.97; 7.0 [5.1]%), SUVmean (0.97; 5.2 [5.8]%), SUVpeak (0.94; 5.3 [4.7]%), TBR (0.96; 14.2 [9.8]%), TMR (0.96; 14.7 [8.4]%), MTV (0.98; 39.9 [41.3]%), and TLH (0.98; 40.1 [43.4]%). There were significant between-scan correlations in all quantitative values. The mean (SD) distance between the two maximum uptake pixels was 7.3 (5.3) mm. CONCLUSIONS We observed a high correlation between the quantitative values at 2 h and 4 h. When using a combined high-quality PET/CT, the total examination time for FMISO-PET can be shortened by skipping the 4-h scan.
Collapse
Affiliation(s)
- Mitsue Kawamura
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Yoichi Shimizu
- Division of Clinical Radiology Service, Kyoto University Hospital, Kyoto, Japan
| | - Kohei Sano
- Division of Clinical Radiology Service, Kyoto University Hospital, Kyoto, Japan
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Mizowaki
- Department of Radiation Oncology and Image-applied Therapy, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Hiraoka
- Department of Radiation Oncology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| |
Collapse
|
18
|
Cancer Detection and Quantification of Treatment Response Using Diffusion-Weighted MRI. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00068-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
19
|
Paterson C, Hargreaves S, Rumley CN. Functional Imaging to Predict Treatment Response in Head and Neck Cancer: How Close are We to Biologically Adaptive Radiotherapy? Clin Oncol (R Coll Radiol) 2020; 32:861-873. [PMID: 33127234 DOI: 10.1016/j.clon.2020.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023]
Abstract
It is increasingly recognised that head and neck cancer represents a spectrum of disease with a differential response to standard treatments. Although prognostic factors are well established, they do not reliably predict response. The ability to predict response early during radiotherapy would allow adaptation of treatment: intensifying treatment for those not responding adequately or de-intensifying remaining therapy for those likely to achieve a complete response. Functional imaging offers such an opportunity. Changes in parameters obtained with functional magnetic resonance imaging or positron emission tomography-computed tomography during treatment have been found to be predictive of disease control in head and neck cancer. Although many questions remain unanswered regarding the optimal implementation of these techniques, current, maturing and future studies may provide the much-needed homogeneous cohorts with larger sample sizes and external validation of parameters. With a stepwise and collaborative approach, we may be able to develop imaging biomarkers that allow us to deliver personalised, biologically adaptive radiotherapy for head and neck cancer.
Collapse
Affiliation(s)
- C Paterson
- Beatson West of Scotland Cancer Centre, Glasgow, UK.
| | | | - C N Rumley
- Department of Radiation Oncology, Townsville University Hospital, Douglas, Australia; South Western Clinical School, University of New South Wales, Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| |
Collapse
|
20
|
Yu W, Su X, Zhang D, Qiao F, Wang H, Jiang J, Xu H. Dual-Tracer Assessment of Dynamic Changes in Reoxygenation and Proliferation Decrease During Fractionated Radiotherapy in Murine Tumors. Front Oncol 2020; 10:1046. [PMID: 32766135 PMCID: PMC7379890 DOI: 10.3389/fonc.2020.01046] [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: 12/12/2019] [Accepted: 05/27/2020] [Indexed: 11/15/2022] Open
Abstract
Objective: The present work aimed to assess reoxygenation and tumor inhibition during fractionated radiotherapy (FRT) in murine tumors using 18F-fluoromisonidazole (18F-FMISO) and 18F-fluorothymidine (18F-FLT) based micro positron emission tomography/computed tomography (PET/CT). Materials and Methods: A nude mouse xenograft model was established with the head and neck squamous carcinoma cell (FaDu), followed by administration of FRT. Imaging was carried out with both 18F-FMISO and 18F-FLT PET/CT, prior to FRT (Pre-FRT, 0 Gy), during FRT (Inter-FRT, 21 Gy), and after FRT (Post-FRT, 40 Gy). The maximum standardized uptake (SUVmax) and tumor-to-normal muscle ratio (TNR) were determined in regions of interest (ROIs) in 18F-FMISO and 18F-FLT PET/CT images. Then, hypoxic (HV) and proliferative tumor (PTV) volumes obtained by PET/CT were analyzed. Immunohistochemistry was performed to analyze the changes of hypoxia-inducible factor- (HIF)-1α, carbonic anhydrase 9 (CAIX), Ki67 and proliferating cell nuclear antigen (PCNA). Associations of the levels of these biomarkers with PET/CT parameters were analyzed. Results:18F-FMISO PET/CT demonstrated markedly elevated reduction rates of SUVmax (30.3 vs. 14.5%, p = 0.012), TNR (27.9 vs. 18.3%, p = 0.032) and HV (85.0 vs. 71.4%, p = 0.047) from Pre-FRT to Inter-FRT compared with values from Inter-FRT to Post-FRT. Meanwhile, PTV reduction rate in 18F-FLT PET/CT from Pre-FRT to Inter-FRT was significantly decreased compared with that from Inter-FRT to Post-FRT (21.2 vs. 82.7%, p = 0.012). Tumor HIF-1α, CAIX, Ki67, and PCNA amounts were continuously down-regulated during radiotherapy. TNR (FMISO) showed significant correlations with HIF-1α (r = 0.692, p = 0.015) and CAIX (r = 0.801, p = 0.006) amounts in xenografts, while associations of SUVmax (FMISO) with hypoxia markers were weak (r = 0.418, p = 0.041 and r = 0.389, p = 0.037, respectively). SUVmax (FLT) was significantly correlated with Ki67 (r = 0.792, p = 0.003) and PCNA (r = 0.837, p = 0.004). Conclusions: Tumor reoxygenation occurs early during radiotherapy, while inhibition of cell proliferation by tumoricidal effects mainly takes place gradually with the course of radiotherapy. 18F-FMISO and 18F-FLT PET/CT are sensitive and non-invasive tools for the monitoring of tumor reoxygenation and proliferation during radiotherapy.
Collapse
Affiliation(s)
- Wenjing Yu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoyu Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dan Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Feng Qiao
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinhui Jiang
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huiqin Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
21
|
Shukla M, Forghani R, Agarwal M. Patient-Centric Head and Neck Cancer Radiation Therapy: Role of Advanced Imaging. Neuroimaging Clin N Am 2020; 30:341-357. [PMID: 32600635 DOI: 10.1016/j.nic.2020.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The traditional 'one-size-fits-all' approach to H&N cancer therapy is archaic. Advanced imaging can identify radioresistant areas by using biomarkers that detect tumor hypoxia, hypercellularity etc. Highly conformal radiotherapy can target resistant areas with precision. The critical information that can be gleaned about tumor biology from these advanced imaging modalities facilitates individualized radiotherapy. The tumor imaging world is pushing its boundaries. Molecular imaging can now detect protein expression and genotypic variations across tumors that can be exploited for tailoring treatment. The exploding field of radiomics and radiogenomics extracts quantitative, biologic and genetic information and further expands the scope of personalized therapy.
Collapse
Affiliation(s)
- Monica Shukla
- Department of Radiation Oncology, Froedtert and Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, WI 53226, USA
| | - Reza Forghani
- Augmented Intelligence & Precision Health Laboratory, Department of Radiology, Research Institute of McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Mohit Agarwal
- Department of Radiology, Section of Neuroradiology, Froedtert and Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
22
|
Wiedenmann N, Grosu AL, Büchert M, Rischke HC, Ruf J, Bielak L, Majerus L, Rühle A, Bamberg F, Baltas D, Hennig J, Mix M, Bock M, Nicolay NH. The utility of multiparametric MRI to characterize hypoxic tumor subvolumes in comparison to FMISO PET/CT. Consequences for diagnosis and chemoradiation treatment planning in head and neck cancer. Radiother Oncol 2020; 150:128-135. [PMID: 32544609 DOI: 10.1016/j.radonc.2020.06.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Hypoxia is an essential metabolic marker that determines chemo- and radiation resistance in head-and-neck squamous cell carcinoma (HNSCC) patients. Our exploratory analysis aimed to identify multiparametric MRI (mpMRI) parameters linked to hypoxia that might be used as surrogate for [18F]FMISO-PET in diagnosis and chemoradiation treatment (CRT) of HNSCC. MATERIALS AND METHODS 21 patients undergoing definitive CRT for HNSCC were prospectively imaged with serial [18F]FMISO-PET and 3 Tesla mpMRI for T1- and T2-weighted and dynamic contrast-enhanced perfusion and diffusion-weighted measurements (ktrans, ve, kep, ADC) in weeks 0, 2 and 5 and FDG-PET in week 0. [18F]FMISO-PET-derived hypoxic subvolumes (HSV) and complementary non-hypoxic subvolumes (nonHSV) were created for tumor and lymph nodes and projected on the mpMRI scans after PET/MRI co-registration. MpMRI and [18F]FMISO-PET parameters within HSVs and nonHSVs were statistically compared. RESULTS FMISO-PET-based HSVs of the primary tumors on MRI were characterized by lower ADC at all time points (p = 0.012 at baseline; p = 0.015 in week 2) and reduced interstitial space volume fraction ve and perfusion ktrans at baseline (p = 0.006, p = 0.047) compared to nonHSVs. Hypoxic lymph nodes were characterized by significantly lower ADC values at baseline (p = 0.039), but not at later time points and a reduction in ktrans-based perfusion at week 2 (p = 0.018). CONCLUSION MpMRI parameters differ significantly between hypoxic and non-hypoxic tumor regions, defined on FMISO-PET/CT as gold standard and might represent surrogate markers for tumor hypoxia. These findings suggest that mpMRI may be useful in the future as a surrogate modality for hypoxia imaging in order to personalize CRT.
Collapse
Affiliation(s)
- Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Büchert
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans C Rischke
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bielak
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Liette Majerus
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Bamberg
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Hennig
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Bock
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
23
|
Chin S, Eccles CL, McWilliam A, Chuter R, Walker E, Whitehurst P, Berresford J, Van Herk M, Hoskin PJ, Choudhury A. Magnetic resonance-guided radiation therapy: A review. J Med Imaging Radiat Oncol 2020; 64:163-177. [PMID: 31646742 DOI: 10.1111/1754-9485.12968] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/24/2019] [Indexed: 12/11/2022]
Abstract
Magnetic resonance-guided radiation therapy (MRgRT) is a promising approach to improving clinical outcomes for patients treated with radiation therapy. The roles of image guidance, adaptive planning and magnetic resonance imaging in radiation therapy have been increasing over the last two decades. Technical advances have led to the feasible combination of magnetic resonance imaging and radiation therapy technologies, leading to improved soft-tissue visualisation, assessment of inter- and intrafraction motion, motion management, online adaptive radiation therapy and the incorporation of functional information into treatment. MRgRT can potentially transform radiation oncology by improving tumour control and quality of life after radiation therapy and increasing convenience of treatment by shortening treatment courses for patients. Multiple groups have developed clinical implementations of MRgRT predominantly in the abdomen and pelvis, with patients having been treated since 2014. While studies of MRgRT have primarily been dosimetric so far, an increasing number of trials are underway examining the potential clinical benefits of MRgRT, with coordinated efforts to rigorously evaluate the benefits of the promising technology. This review discusses the current implementations, studies, potential benefits and challenges of MRgRT.
Collapse
Affiliation(s)
- Stephen Chin
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Westmead Clinical School, University of Sydney, Sydney, New South Wales, Australia
| | - Cynthia L Eccles
- Department of Radiotherapy, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Alan McWilliam
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Robert Chuter
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Emma Walker
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Philip Whitehurst
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Joseph Berresford
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Marcel Van Herk
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, UK
| | - Peter J Hoskin
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Ananya Choudhury
- Department of Clinical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
24
|
Deschuymer S, Nevens D, Duprez F, Daisne JF, Dok R, Laenen A, Voordeckers M, De Neve W, Nuyts S. Randomized clinical trial on reduction of radiotherapy dose to the elective neck in head and neck squamous cell carcinoma; update of the long-term tumor outcome. Radiother Oncol 2020; 143:24-29. [DOI: 10.1016/j.radonc.2020.01.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/06/2020] [Accepted: 01/07/2020] [Indexed: 01/13/2023]
|
25
|
Atwell D, Elks J, Cahill K, Hearn N, Vignarajah D, Lagopoulos J, Min M. A Review of Modern Radiation Therapy Dose Escalation in Locally Advanced Head and Neck Cancer. Clin Oncol (R Coll Radiol) 2020; 32:330-341. [PMID: 31911016 DOI: 10.1016/j.clon.2019.12.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/27/2019] [Accepted: 11/07/2019] [Indexed: 12/30/2022]
Abstract
The management of head and neck cancer is complex and often involves multimodality treatment. Certain groups of patients, such as those with inoperable or advanced disease, are at higher risk of treatment failure and may therefore benefit from radiation therapy dose escalation. This can be difficult to achieve without increasing toxicity. However, the combination of modern treatment techniques and increased research into the use of functional imaging modalities that assist with target delineation allows researchers to push this boundary further. This review aims to summarise modern dose escalation trials to identify the impact on disease outcomes and explore the growing role of functional imaging modalities. Studies experimenting with dose escalation above standard fractionated regimens as outlined in National Comprehensive Cancer Network guidelines using photon therapy were chosen for review. Seventeen papers were considered suitable for inclusion in the review. Eight studies investigated nasopharyngeal cancer, with the remainder treating a range of subsites. Six studies utilised functional imaging modalities for target delineation. Doses as high as 85.9 Gy in 2.6 Gy fractions (EQD2 90.2 Gy10) were reportedly delivered with the aid of functional imaging modalities. Dose escalation in nasopharyngeal cancer resulted in 3-year locoregional control rates of 86.6-100% and overall survival of 82-95.2%. For other mucosal primary tumour sites, 3-year locoregional control reached 68.2-85.9% and 48.4-54% for overall survival. There were no clear trends in acute or late toxicity across studies, regardless of dose or addition of chemotherapy. However, small cohort sizes and short follow-up times may have resulted in under-reporting. This review highlights the future possibilities of radiation therapy dose escalation in head and neck cancer and the potential for improved target delineation with careful patient selection and the assistance of functional imaging modalities.
Collapse
Affiliation(s)
- D Atwell
- Cancer Services, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; Icon Cancer Centre, Maroochydore, Queensland, Australia; Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia.
| | - J Elks
- Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - K Cahill
- Cancer Services, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - N Hearn
- Cancer Services, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; Icon Cancer Centre, Maroochydore, Queensland, Australia; Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - D Vignarajah
- Cancer Services, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; Icon Cancer Centre, Maroochydore, Queensland, Australia
| | - J Lagopoulos
- Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - M Min
- Cancer Services, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; Icon Cancer Centre, Maroochydore, Queensland, Australia; Sunshine Coast Mind and Neuroscience - Thompson Institute, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| |
Collapse
|
26
|
Beaumont J, Acosta O, Devillers A, Palard-Novello X, Chajon E, de Crevoisier R, Castelli J. Voxel-based identification of local recurrence sub-regions from pre-treatment PET/CT for locally advanced head and neck cancers. EJNMMI Res 2019; 9:90. [PMID: 31535233 PMCID: PMC6751236 DOI: 10.1186/s13550-019-0556-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Background Overall, 40% of patients with a locally advanced head and neck cancer (LAHNC) treated by chemoradiotherapy (CRT) present local recurrence within 2 years after the treatment. The aims of this study were to characterize voxel-wise the sub-regions where tumor recurrence appear and to predict their location from pre-treatment 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) images. Materials and methods Twenty-six patients with local failure after treatment were included in this study. Local recurrence volume was identified by co-registering pre-treatment and recurrent PET/CT images using a customized rigid registration algorithm. A large set of voxel-wise features were extracted from pre-treatment PET to train a random forest model allowing to predict local recurrence at the voxel level. Results Out of 26 expert-assessed registrations, 15 provided enough accuracy to identify recurrence volumes and were included for further analysis. Recurrence volume represented on average 23% of the initial tumor volume. The MTV with a threshold of 50% of SUVmax plus a 3D margin of 10 mm covered on average 89.8% of the recurrence and 96.9% of the initial tumor. SUV and MTV alone were not sufficient to identify the area of recurrence. Using a random forest model, 15 parameters, combining radiomics and spatial location, were identified, allowing to predict the recurrence sub-regions with a median area under the receiver operating curve of 0.71 (range 0.14–0.91). Conclusion As opposed to regional comparisons which do not bring enough evidence for accurate prediction of recurrence volume, a voxel-wise analysis of FDG-uptake features suggested a potential to predict recurrence with enough accuracy to consider tailoring CRT by dose escalation within likely radioresistant regions.
Collapse
Affiliation(s)
- J Beaumont
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France
| | - O Acosta
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France
| | - A Devillers
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France.,Department of Radiotherapy, Centre Eugene Marquis, avenue de la Bataille Flandre Dunkerque, 35000, Rennes, France
| | - X Palard-Novello
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France.,Department of Radiotherapy, Centre Eugene Marquis, avenue de la Bataille Flandre Dunkerque, 35000, Rennes, France
| | - E Chajon
- Department of Radiotherapy, Centre Eugene Marquis, avenue de la Bataille Flandre Dunkerque, 35000, Rennes, France
| | - R de Crevoisier
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France.,Department of Radiotherapy, Centre Eugene Marquis, avenue de la Bataille Flandre Dunkerque, 35000, Rennes, France
| | - J Castelli
- Univ Rennes, CLCC Eugène Marquis, INSERM, LTSI - UMR 1099, 35000, Rennes, France. .,Department of Radiotherapy, Centre Eugene Marquis, avenue de la Bataille Flandre Dunkerque, 35000, Rennes, France.
| |
Collapse
|
27
|
Stieb S, McDonald B, Gronberg M, Engeseth GM, He R, Fuller CD. Imaging for Target Delineation and Treatment Planning in Radiation Oncology: Current and Emerging Techniques. Hematol Oncol Clin North Am 2019; 33:963-975. [PMID: 31668214 DOI: 10.1016/j.hoc.2019.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Imaging in radiation oncology has a wide range of applications. It is necessary not only for tumor staging and treatment response assessment after therapy but also for the treatment planning process, including definition of target and organs at risk, as well as treatment plan calculation. This article provides a comprehensive overview of the main imaging modalities currently used for target delineation and treatment planning and gives insight into new and promising techniques.
Collapse
Affiliation(s)
- Sonja Stieb
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Brigid McDonald
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Mary Gronberg
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Grete May Engeseth
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Renjie He
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Clifton David Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
28
|
PET and MRI based RT treatment planning: Handling uncertainties. Cancer Radiother 2019; 23:753-760. [PMID: 31427076 DOI: 10.1016/j.canrad.2019.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022]
Abstract
Imaging provides the basis for radiotherapy. Multi-modality images are used for target delineation (primary tumor and nodes, boost volume) and organs at risk, treatment guidance, outcome prediction, and treatment assessment. Next to anatomical information, more and more functional imaging is being used. The current paper provides a brief overview of the different applications of imaging techniques used in the radiotherapy process, focusing on uncertainties and QA. The paper mainly focuses on PET and MRI, but also provides a short discussion on DCE-CT. A close collaboration between radiology, nuclear medicine and radiotherapy departments provides the key to improve the quality of radiotherapy. Jointly developed imaging protocols (RT position setup, immobilization tools, lasers, flat table…), and QA programs are mandatory. For PET, suitable windowing in consultation with a Nuclear Medicine Physician is crucial (differentiation benign/malignant lesions, artifacts…). A basic knowledge of MRI sequences is required, in such a way that geometrical distortions are easily recognized by all members the RT and RT physics team. If this is not the case, then the radiologist should be introduced systematically in the delineation process and multidisciplinary meetings need to be organized regularly. For each image modality and each image registration process, the associated uncertainties need to be determined and integrated in the PTV margin. When using functional information for dose painting, response assessment or outcome prediction, collaboration between the different departments is even more important. Limitations of imaging based biomarkers (specificity, sensitivity) should be known.
Collapse
|
29
|
Integrating molecular nuclear imaging in clinical research to improve anticancer therapy. Nat Rev Clin Oncol 2019; 16:241-255. [PMID: 30479378 DOI: 10.1038/s41571-018-0123-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective patient selection before or early during treatment is important to increasing the therapeutic benefits of anticancer treatments. This selection process is often predicated on biomarkers, predominantly biospecimen biomarkers derived from blood or tumour tissue; however, such biomarkers provide limited information about the true extent of disease or about the characteristics of different, potentially heterogeneous tumours present in an individual patient. Molecular imaging can also produce quantitative outputs; such imaging biomarkers can help to fill these knowledge gaps by providing complementary information on tumour characteristics, including heterogeneity and the microenvironment, as well as on pharmacokinetic parameters, drug-target engagement and responses to treatment. This integrative approach could therefore streamline biomarker and drug development, although a range of issues need to be overcome in order to enable a broader use of molecular imaging in clinical trials. In this Perspective article, we outline the multistage process of developing novel molecular imaging biomarkers. We discuss the challenges that have restricted the use of molecular imaging in clinical oncology research to date and outline future opportunities in this area.
Collapse
|
30
|
Gérard M, Corroyer-Dulmont A, Lesueur P, Collet S, Chérel M, Bourgeois M, Stefan D, Limkin EJ, Perrio C, Guillamo JS, Dubray B, Bernaudin M, Thariat J, Valable S. Hypoxia Imaging and Adaptive Radiotherapy: A State-of-the-Art Approach in the Management of Glioma. Front Med (Lausanne) 2019; 6:117. [PMID: 31249831 PMCID: PMC6582242 DOI: 10.3389/fmed.2019.00117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/13/2019] [Indexed: 01/31/2023] Open
Abstract
Severe hypoxia [oxygen partial pressure (pO2) below 5–10 mmHg] is more frequent in glioblastoma multiforme (GBM) compared to lower-grade gliomas. Seminal studies in the 1950s demonstrated that hypoxia was associated with increased resistance to low–linear energy transfer (LET) ionizing radiation. In experimental conditions, the total radiation dose has to be multiplied by a factor of 3 to achieve the same cell lethality in anoxic situations. The presence of hypoxia in human tumors is assumed to contribute to treatment failures after radiotherapy (RT) in cancer patients. Therefore, a logical way to overcome hypoxia-induced radioresistance would be to deliver substantially higher doses of RT in hypoxic volumes delineated on pre-treatment imaging as biological target volumes (BTVs). Such an approach faces various fundamental, technical, and clinical challenges. The present review addresses several technical points related to the delineation of hypoxic zones, which include: spatial accuracy, quantitative vs. relative threshold, variations of hypoxia levels during RT, and availability of hypoxia tracers. The feasibility of hypoxia imaging as an assessment tool for early tumor response to RT and for predicting long-term outcomes is discussed. Hypoxia imaging for RT dose painting is likewise examined. As for the radiation oncologist's point of view, hypoxia maps should be converted into dose-distribution objectives for RT planning. Taking into account the physics and the radiobiology of various irradiation beams, preliminary in silico studies are required to investigate the feasibility of dose escalation in terms of normal tissue tolerance before clinical trials are undertaken.
Collapse
Affiliation(s)
- Michael Gérard
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | | | - Paul Lesueur
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Solène Collet
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiophysics, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Michel Chérel
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Mickael Bourgeois
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Dinu Stefan
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Elaine Johanna Limkin
- Department of Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Cécile Perrio
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Jean-Sébastien Guillamo
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Neurology, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Bernard Dubray
- Département de Radiothérapie et de Physique Médicale, Laboratoire QuantIF-LITIS [EA 4108], Centre de Lutte Contre le Cancer Henri Becquerel, Université de Normandie, Rouen, France
| | - Myriam Bernaudin
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Juliette Thariat
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Samuel Valable
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| |
Collapse
|
31
|
Beaton L, Bandula S, Gaze MN, Sharma RA. How rapid advances in imaging are defining the future of precision radiation oncology. Br J Cancer 2019; 120:779-790. [PMID: 30911090 PMCID: PMC6474267 DOI: 10.1038/s41416-019-0412-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/23/2019] [Accepted: 02/05/2019] [Indexed: 12/13/2022] Open
Abstract
Imaging has an essential role in the planning and delivery of radiotherapy. Recent advances in imaging have led to the development of advanced radiotherapy techniques—including image-guided radiotherapy, intensity-modulated radiotherapy, stereotactic body radiotherapy and proton beam therapy. The optimal use of imaging might enable higher doses of radiation to be delivered to the tumour, while sparing normal surrounding tissues. In this article, we review how the integration of existing and novel forms of computed tomography, magnetic resonance imaging and positron emission tomography have transformed tumour delineation in the radiotherapy planning process, and how these advances have the potential to allow a more individualised approach to the cancer therapy. Recent data suggest that imaging biomarkers that assess underlying tumour heterogeneity can identify areas within a tumour that are at higher risk of radio-resistance, and therefore potentially allow for biologically focussed dose escalation. The rapidly evolving concept of adaptive radiotherapy, including artificial intelligence, requires imaging during treatment to be used to modify radiotherapy on a daily basis. These advances have the potential to improve clinical outcomes and reduce radiation-related long-term toxicities. We outline how recent technological advances in both imaging and radiotherapy delivery can be combined to shape the future of precision radiation oncology.
Collapse
Affiliation(s)
- Laura Beaton
- Cancer Institute, University College London, London, UK
| | - Steve Bandula
- Cancer Institute, University College London, London, UK.,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Mark N Gaze
- NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK
| | - Ricky A Sharma
- Cancer Institute, University College London, London, UK. .,NIHR University College London Hospitals Biomedical Research Centre, UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
32
|
Eckert F, Zwirner K, Boeke S, Thorwarth D, Zips D, Huber SM. Rationale for Combining Radiotherapy and Immune Checkpoint Inhibition for Patients With Hypoxic Tumors. Front Immunol 2019; 10:407. [PMID: 30930892 PMCID: PMC6423917 DOI: 10.3389/fimmu.2019.00407] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022] Open
Abstract
In order to compensate for the increased oxygen consumption in growing tumors, tumors need angiogenesis and vasculogenesis to increase the supply. Insufficiency in this process or in the microcirculation leads to hypoxic tumor areas with a significantly reduced pO2, which in turn leads to alterations in the biology of cancer cells as well as in the tumor microenvironment. Cancer cells develop more aggressive phenotypes, stem cell features and are more prone to metastasis formation and migration. In addition, intratumoral hypoxia confers therapy resistance, specifically radioresistance. Reactive oxygen species are crucial in fixing DNA breaks after ionizing radiation. Thus, hypoxic tumor cells show a two- to threefold increase in radioresistance. The microenvironment is enriched with chemokines (e.g., SDF-1) and growth factors (e.g., TGFβ) additionally reducing radiosensitivity. During recent years hypoxia has also been identified as a major factor for immune suppression in the tumor microenvironment. Hypoxic tumors show increased numbers of myeloid derived suppressor cells (MDSCs) as well as regulatory T cells (Tregs) and decreased infiltration and activation of cytotoxic T cells. The combination of radiotherapy with immune checkpoint inhibition is on the rise in the treatment of metastatic cancer patients, but is also tested in multiple curative treatment settings. There is a strong rationale for synergistic effects, such as increased T cell infiltration in irradiated tumors and mitigation of radiation-induced immunosuppressive mechanisms such as PD-L1 upregulation by immune checkpoint inhibition. Given the worse prognosis of patients with hypoxic tumors due to local therapy resistance but also increased rate of distant metastases and the strong immune suppression induced by hypoxia, we hypothesize that the subgroup of patients with hypoxic tumors might be of special interest for combining immune checkpoint inhibition with radiotherapy.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Zwirner
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Simon Boeke
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
33
|
Repeat FMISO-PET imaging weakly correlates with hypoxia-associated gene expressions for locally advanced HNSCC treated by primary radiochemotherapy. Radiother Oncol 2019; 135:43-50. [PMID: 31015169 DOI: 10.1016/j.radonc.2019.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/07/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hypoxia is an important factor of tumour resistance to radiotherapy, chemotherapy and potentially immunotherapy. It can be measured e.g. by positron emission tomography (PET) imaging or hypoxia-associated gene expressions from tumour biopsies. Here we correlate [18F]fluoromisonidazole (FMISO)-PET/CT imaging with hypoxia-associated gene expressions on a cohort of 50 head and neck squamous cell carcinoma (HNSCC) patients and compare their prognostic value for response to radiochemotherapy (RCTx). METHODS FMISO-PET/CT images of 50 HNSCC patients were acquired at four time-points before and during RCTx. For 42 of these patients, hypoxia-associated gene expressions were evaluated by nanoString technology based on a biopsy obtained before any treatment. The FMISO-PET parameters tumour-to-background ratio and hypoxic volume were correlated to the expressions of 58 hypoxia-associated genes using the Spearman correlation coefficient ρ. Three hypoxia-associated gene signatures were compared regarding their correlation with the FMISO-PET parameters using their median expression. In addition, the correlation with tumour volume was analysed. The impact of both hypoxia measurement methods on loco-regional tumour control (LRC) and overall survival (OS) was assessed by Cox regression. RESULTS The median expression of hypoxia-associated genes was weakly correlated to hypoxia measured by FMISO-PET imaging (ρ ≤ 0.43), with higher correlations to imaging after weeks 1 and 2 of treatment (p < 0.001). Moderate correlations were obtained between FMISO-PET imaging and tumour volume (ρ ≤ 0.69). Prognostic models for LRC and OS based on the FMISO-PET parameters could not be improved by including hypoxia classifiers. CONCLUSION We observed low correlations between hypoxia FMISO-PET parameters and expressions of hypoxia-associated genes. Since FMISO-PET showed a superior patient stratification, it may be the preferred biomarker over hypoxia-associated genes for stratifying patients with locally advanced HNSCC treated by primary RCTx.
Collapse
|
34
|
Bandurska-Luque A, Löck S, Haase R, Richter C, Zöphel K, Perrin R, Appold S, Krause M, Steinbach J, Kotzerke J, Hofheinz F, Zips D, Baumann M, Troost EG. Correlation between FMISO-PET based hypoxia in the primary tumour and in lymph node metastases in locally advanced HNSCC patients. Clin Transl Radiat Oncol 2019; 15:108-112. [PMID: 30834349 PMCID: PMC6384311 DOI: 10.1016/j.ctro.2019.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
We investigated correlation between hypoxia in the primary tumour and LN before and during RCTx. The Correlation between primary tumour and LN hypoxia is stronger in patients with large LN compared to the entire cohort. We advise to perform a comprehensive evaluation of hypoxia in the primary tumour and LN.
Purpose This secondary analysis of the prospective study on repeat [18F]fluoromisonidazole (FMISO)-PET in patients with locally advanced head and neck squamous cell carcinoma (HNSCC) assessed the correlation of hypoxia in the primary tumour and lymph node metastases (LN) prior to and during primary radiochemotherapy. Methods This analysis included forty-five LN-positive HNSCC patients having undergone FMISO-PET/CTs at baseline, and at week 1, 2 and 5 of radiochemotherapy. The quantitative FMISO-PET/CT parameters maximum standardised uptake value (SUVmax, corrected for partial volume effect) and peak tumour-to-background ratio (TBRpeak) were estimated in the primary tumour as well as in index and large LN, respectively. Statistical analysis was performed using the Spearman correlation coefficient ρ. Results In 15 patients with large LN (FDG-PET positive volume >5 ml), there was a significant correlation between the hypoxia measured in the primary tumour and the large LN at three out of four time-points using the TBRpeak (baseline: ρ = 0.57, p = 0.006; week 2: ρ = 0.64, p = 0.003 and week 5: ρ = 0.68, p = 0.001). For the entire cohort (N = 45) only assessed prior to the treatment, there was a statistically significant, though weak correlation between FMISO-SUVmax of the primary tumour and the index LN (ρ = 0.36, p = 0.015). Conclusions We observed a significant correlation between FMISO-based hypoxia in the primary tumour and large lymph node(s) in advanced stage HNSCC patients. However, since most patients only had relatively small hypoxic lymph node metastases, a comprehensive assessment of the primary tumour and lymph node hypoxia is essential.
Collapse
Affiliation(s)
- Anna Bandurska-Luque
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Corresponding author at: Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus Dresden, Fetscherstrasse 74, 01307 Dresden, 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, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Haase
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Christian Richter
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 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
| | - Klaus Zöphel
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- National Center for Tumor Diseases, 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; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Rosalind Perrin
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Steffen Appold
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, 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, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 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, 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; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Rossendorf, Germany
- Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Frank Hofheinz
- Helmholtz-Zentrum Dresden-Rossendorf, PET Center, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls Universität Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, 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, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases, 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; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Esther G.C. Troost
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 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, 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; Helmholtz Association / Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| |
Collapse
|
35
|
Martens RM, Noij DP, Ali M, Koopman T, Marcus JT, Vergeer MR, de Vet H, de Jong MC, Leemans CR, Hoekstra OS, de Bree R, de Graaf P, Boellaard R, Castelijns JA. Functional imaging early during (chemo)radiotherapy for response prediction in head and neck squamous cell carcinoma; a systematic review. Oral Oncol 2018; 88:75-83. [PMID: 30616800 DOI: 10.1016/j.oraloncology.2018.11.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 10/27/2022]
Abstract
This systematic review gives an extensive overview of the current state of functional imaging during (chemo)radiotherapy to predict locoregional control (LRC) and overall survival (OS) for head and neck squamous cell carcinoma. MEDLINE and EMBASE were searched for literature until April 2018 assessing the predictive performance of functional imaging (computed tomography perfusion (CTp), MRI and positron-emission tomography (PET)) within 4 weeks after (chemo)radiotherapy initiation. Fifty-two studies (CTp: n = 4, MRI: n = 19, PET: n = 26, MRI/PET: n = 3) were included involving 1623 patients. Prognostic information was extracted according the PRISMA protocol. Pooled estimation and subgroup analyses were performed for comparable parameters and outcome. However, the heterogeneity of included studies limited the possibility for comparison. Early tumoral changes from (chemo)radiotherapy can be captured by functional MRI and 18F-FDG-PET and could allow for personalized treatment adaptation. Lesions showed potentially prognostic intratreatment changes in perfusion, diffusion and metabolic activity. Intratreatment ADCmean increase (decrease of diffusion restriction) and low SUVmax (persistent low or decrease of 18F-FDG uptake) were most predictive of LRC. Intratreatment persistent high or increase of perfusion on CT/MRI (i.e. blood flow, volume, permeability) also predicted LRC. Low SUVmax and total lesion glycolysis (TLG) predicted favorable OS. The optimal timing to perform functional imaging to predict LRC or OS was 2-3 weeks after treatment initiation.
Collapse
Affiliation(s)
- Roland M Martens
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands.
| | - Daniel P Noij
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Meedie Ali
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Thomas Koopman
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - J Tim Marcus
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Marije R Vergeer
- Department of Radiation Oncology, VU University Medical Center, Amsterdam, the Netherlands
| | - Henrica de Vet
- Department of Epidemiology and Biostatistics and the EMGO Institute for Health and Care Research, VU University Medical Center, Amsterdam, the Netherlands
| | - Marcus C de Jong
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - C René Leemans
- Department of Otolaryngology - Head and Neck Surgery, VU University Medical Center, Amsterdam, the Netherlands
| | - Otto S Hoekstra
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Pim de Graaf
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| | - Jonas A Castelijns
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, the Netherlands
| |
Collapse
|
36
|
Dual-energy computed tomography for prediction of loco-regional recurrence after radiotherapy in larynx and hypopharynx squamous cell carcinoma. Eur J Radiol 2018; 110:1-6. [PMID: 30599844 DOI: 10.1016/j.ejrad.2018.11.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 10/28/2018] [Accepted: 11/04/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE To investigate the role of quantitative pre-treatment dual-energy computed tomography (DECT) for prediction of loco-regional recurrence (LRR) in patients with larynx/hypopharynx squamous cell cancer (L/H SCC). METHODS Patients with L/H SCC treated with curative intent loco-regional radiotherapy and that underwent treatment planning with contrast-enhanced DECT of the neck were included. Primary and nodal gross tumor volumes (GTVp and GTVn) were contoured and transferred into a Matlab® workspace. Using a two-material decomposition, GTV iodine concentration (IC) maps were obtained. Quantitative histogram statistics (maximum, mean, standard deviation, kurtosis and skewness) were retrieved from the IC maps. Cox regression analysis was conducted to determine potential predictive factors of LRR. RESULTS Twenty-five patients, including 20 supraglottic and 5 pyriform sinus tumors were analysed. Stage I, II, III, IVa and IVb constituted 4% (1 patient), 24%, 36%, 28% and 8% of patients, respectively; 44% had concurrent chemo-radiotherapy and 28% had neodjuvant chemotherapy. Median follow-up was 21 months. Locoregional control at 1 and 2 years were 75% and 69%, respectively. For the entire cohort, GTVn volume (HR 1.177 [1.001-1.392], p = 0.05), voxel-based maximum IC of GTVp (HR 1.099 [95% CI: 1.001-1.209], p = 0.05) and IC standard deviation of GTVn (HR 9.300 [95% CI: 1.113-77.725] p = 0.04) were predictive of LRR. On subgroup analysis of patients treated with upfront radiotherapy +/- chemotherapy, both voxel-based maximum IC of GTVp (HR 1.127 [95% CI: 1.010-1.258], p = 0.05) and IC kurtosis of GTVp (HR 1.088 [95% CI: 1.014-1.166], p = 0.02) were predictive of LRR. CONCLUSION This exploratory study suggests that pre-radiotherapy DECT-derived IC quantitative analysis of tumoral volume may help predict LRR in L/H SCC.
Collapse
|
37
|
Hamming-Vrieze O, Navran A, Al-Mamgani A, Vogel WV. Biological PET-guided adaptive radiotherapy for dose escalation in head and neck cancer: a systematic review. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF RADIOPHARMACEUTICAL CHEMISTRY AND BIOLOGY 2018; 62:349-368. [DOI: 10.23736/s1824-4785.18.03087-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
38
|
Gensheimer MF, Le QT. Adaptive radiotherapy for head and neck cancer: Are we ready to put it into routine clinical practice? Oral Oncol 2018; 86:19-24. [DOI: 10.1016/j.oraloncology.2018.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
|
39
|
FMISO-PET-based lymph node hypoxia adds to the prognostic value of tumor only hypoxia in HNSCC patients. Radiother Oncol 2018; 130:97-103. [PMID: 30293643 DOI: 10.1016/j.radonc.2018.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 11/23/2022]
Abstract
PURPOSE This secondary analysis of the prospective study on repeat [18F]fluoromisonidazole (FMISO)-PET in patients with locally advanced head and neck squamous cell carcinomas (HNSCC) assessed the prognostic value of synchronous hypoxia in primary tumor (Tu) and lymph node metastases (LN), and evaluated whether the combined reading was of higher prognostic value than that of primary tumor hypoxia only. METHODS This analysis included forty-five LN-positive HNSCC patients. FMISO-PET/CTs were performed at baseline, weeks 1, 2 and 5 of radiochemotherapy. Based on a binary scale, Tu and LN were categorized as hypoxic or normoxic, and two prognostic parameters were defined: Tu-hypoxia (independent of the LN oxygenation status) and synchronous Tu-and-LN-hypoxia. In fifteen patients with large LN (N = 21), additional quantitative analyses of FMISO-PET/CTs were performed. Imaging parameters at different time-points were correlated to the endpoints, i.e., locoregional control (LRC), local control (LC), regional control (RC) and time to progression (TTP). Survival curves were estimated using the cumulative incidence function. Univariable and multivariable Cox regression was used to evaluate the prognostic impact of hypoxia on the endpoints. RESULTS Synchronous Tu-and-LN-hypoxia was a strong adverse prognostic factor for LC, LRC and TTP at any of the four time-points (p ≤ 0.004), whereas Tu-hypoxia only was significantly associated with poor LC and LRC in weeks 2 and 5 (p ≤ 0.047), and with TTP in week 1 (p = 0.046). The multivariable analysis confirmed the prognostic value of synchronous Tu-and-LN-hypoxia regarding LRC (HR = 14.8, p = 0.017). The quantitative FMISO-PET/CT parameters correlated with qualitative hypoxia scale and RC (p < 0.001, p ≤ 0.033 at week 2, respectively). CONCLUSIONS This secondary analysis suggests that combined reading of primary tumor and LN hypoxia adds to the prognostic information of FMSIO-PET in comparison to primary tumor assessment alone in particular prior and early during radiochemotherapy. Confirmation in ongoing trials is needed before using this marker for personalized radiation oncology.
Collapse
|
40
|
Bonnitcha P, Grieve S, Figtree G. Clinical imaging of hypoxia: Current status and future directions. Free Radic Biol Med 2018; 126:296-312. [PMID: 30130569 DOI: 10.1016/j.freeradbiomed.2018.08.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/20/2022]
Abstract
Tissue hypoxia is a key feature of many important causes of morbidity and mortality. In pathologies such as stroke, peripheral vascular disease and ischaemic heart disease, hypoxia is largely a consequence of low blood flow induced ischaemia, hence perfusion imaging is often used as a surrogate for hypoxia to guide clinical diagnosis and treatment. Importantly, ischaemia and hypoxia are not synonymous conditions as it is not universally true that well perfused tissues are normoxic or that poorly perfused tissues are hypoxic. In pathologies such as cancer, for instance, perfusion imaging and oxygen concentration are less well correlated, and oxygen concentration is independently correlated to radiotherapy response and overall treatment outcomes. In addition, the progression of many diseases is intricately related to maladaptive responses to the hypoxia itself. Thus there is potentially great clinical and scientific utility in direct measurements of tissue oxygenation. Despite this, imaging assessment of hypoxia in patients is rarely performed in clinical settings. This review summarises some of the current methods used to clinically evaluate hypoxia, the barriers to the routine use of these methods and the newer agents and techniques being explored for the assessment of hypoxia in pathological processes.
Collapse
Affiliation(s)
- Paul Bonnitcha
- Northern and Central Clinical Schools, Faculty of Medicine, Sydney University, Sydney, NSW 2006, Australia; Chemical Pathology Department, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia.
| | - Stuart Grieve
- Sydney Translational Imaging Laboratory, Heart Research Institute, Charles Perkins Centre and Sydney Medical School, University of Sydney, NSW 2050, Australia
| | - Gemma Figtree
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia; Cardiology Department, Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia
| |
Collapse
|
41
|
Leibfarth S, Winter RM, Lyng H, Zips D, Thorwarth D. Potentials and challenges of diffusion-weighted magnetic resonance imaging in radiotherapy. Clin Transl Radiat Oncol 2018; 13:29-37. [PMID: 30294681 PMCID: PMC6169338 DOI: 10.1016/j.ctro.2018.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/20/2018] [Accepted: 09/03/2018] [Indexed: 02/09/2023] Open
Abstract
Discussion of DW imaging protocols and imaging setup. Discussion of mono- and bi-exponential models for quantitative parameter extraction. Review of recent publications investigating potential benefits of using DWI in RT, including detailed synoptic table. Detailed discussion of geometric and quantitative accuracy of DW imaging and DW-derived parameters.
Purpose To review the potential and challenges of integrating diffusion weighted magnetic resonance imaging (DWI) into radiotherapy (RT). Content Details related to image acquisition of DWI for RT purposes are discussed, along with the challenges with respect to geometric accuracy and the robustness of quantitative parameter extraction. An overview of diffusion- and perfusion-related parameters derived from mono- and bi-exponential models is provided, and their role as potential RT biomarkers is discussed. Recent studies demonstrating potential of DWI in different tumor sites such as the head and neck, rectum, cervix, prostate, and brain, are reviewed in detail. Conclusion DWI has shown promise for RT outcome prediction, response assessment, as well as for tumor delineation and characterization in several cancer types. Geometric and quantification robustness is challenging and has to be addressed adequately. Evaluation in larger clinical trials with well designed imaging protocol and advanced analysis models is needed to develop the optimal strategy for integrating DWI in RT.
Collapse
Affiliation(s)
- Sara Leibfarth
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Germany
| | - René M Winter
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Germany
| | - Heidi Lyng
- Department of Radiation Biology, Norwegian Radium Hospital, Oslo University Hospital, Norway
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, Germany
| | - Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Germany
| |
Collapse
|
42
|
Winter RM, Leibfarth S, Schmidt H, Zwirner K, Mönnich D, Welz S, Schwenzer NF, la Fougère C, Nikolaou K, Gatidis S, Zips D, Thorwarth D. Assessment of image quality of a radiotherapy-specific hardware solution for PET/MRI in head and neck cancer patients. Radiother Oncol 2018; 128:485-491. [PMID: 29747873 PMCID: PMC6141811 DOI: 10.1016/j.radonc.2018.04.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/29/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND PURPOSE Functional PET/MRI has great potential to improve radiotherapy planning (RTP). However, data integration requires imaging with radiotherapy-specific patient positioning. Here, we investigated the feasibility and image quality of radiotherapy-customized PET/MRI in head-and-neck cancer (HNC) patients using a dedicated hardware setup. MATERIAL AND METHODS Ten HNC patients were examined with simultaneous PET/MRI before treatment, with radiotherapy and diagnostic scan setup, respectively. We tested feasibility of radiotherapy-specific patient positioning and compared the image quality between both setups by pairwise image analysis of 18F-FDG-PET, T1/T2-weighted and diffusion-weighted MRI. For image quality assessment, similarity measures including average symmetric surface distance (ASSD) of PET and MR-based tumor contours, MR signal-to-noise ratio (SNR) and mean apparent diffusion coefficient (ADC) value were used. RESULTS PET/MRI in radiotherapy position was feasible - all patients were successfully examined. ASSD (median/range) of PET and MR contours was 0.6 (0.4-1.2) and 0.9 (0.5-1.3) mm, respectively. For T2-weighted MRI, a reduced SNR of -26.2% (-39.0--11.7) was observed with radiotherapy setup. No significant difference in mean ADC was found. CONCLUSIONS Simultaneous PET/MRI in HNC patients using radiotherapy positioning aids is clinically feasible. Though SNR was reduced, the image quality obtained with a radiotherapy setup meets RTP requirements and the data can thus be used for personalized RTP.
Collapse
Affiliation(s)
- René M Winter
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany.
| | - Sara Leibfarth
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - Holger Schmidt
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - Kerstin Zwirner
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - David Mönnich
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Welz
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - Nina F Schwenzer
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - Christian la Fougère
- Department of Nuclear Medicine, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sergios Gatidis
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
43
|
Stieb S, Eleftheriou A, Warnock G, Guckenberger M, Riesterer O. Longitudinal PET imaging of tumor hypoxia during the course of radiotherapy. Eur J Nucl Med Mol Imaging 2018; 45:2201-2217. [PMID: 30128659 DOI: 10.1007/s00259-018-4116-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022]
Abstract
Hypoxia results from an imbalance between oxygen supply and consumption. It is a common phenomenon in solid malignant tumors such as head and neck cancer. As hypoxic cells are more resistant to therapy, tumor hypoxia is an indicator for poor prognosis. Several techniques have been developed to measure tissue oxygenation. These are the Eppendorf O2 polarographic needle electrode, immunohistochemical analysis of endogenous (e.g., hypoxia-inducible factor-1α (HIF-1a)) and exogenous markers (e.g., pimonidazole) as well as imaging methods such as functional magnetic resonance imaging (e.g., blood oxygen level dependent (BOLD) imaging, T1-weighted imaging) and hypoxia positron emission tomography (PET). Among the imaging modalities, only PET is sufficiently validated to detect hypoxia for clinical use. Hypoxia PET tracers include 18F-fluoromisonidazole (FMISO), the most commonly used hypoxic marker, 18F-flouroazomycin arabinoside (FAZA), 18Ffluoroerythronitroimidazole (FETNIM), 18F-2-nitroimidazolpentafluoropropylacetamide (EF5) and 18F-flortanidazole (HX4). As technical development provides the opportunity to increase the radiation dose to subregions of the tumor, such as hypoxic areas, it has to be ensured that these regions are stable not only from imaging to treatment but also through the course of radiotherapy. The aim of this review is therefore to characterize the behavior of tumor hypoxia during radiotherapy for the whole tumor and for subregions by using hypoxia PET tracers, with focus on head and neck cancer patients.
Collapse
Affiliation(s)
- Sonja Stieb
- Department of Radiation Oncology, University Hospital and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland. .,Institute of Diagnostic and Interventional Radiology, University Hospital and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
| | - Afroditi Eleftheriou
- Department of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Geoffrey Warnock
- Department of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,Department of Nuclear Medicine, University Hospital and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Oliver Riesterer
- Department of Radiation Oncology, University Hospital and University of Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| |
Collapse
|
44
|
Kelada OJ, Rockwell S, Zheng MQ, Huang Y, Liu Y, Booth CJ, Decker RH, Oelfke U, Carson RE, Carlson DJ. Quantification of Tumor Hypoxic Fractions Using Positron Emission Tomography with [ 18F]Fluoromisonidazole ([ 18F]FMISO) Kinetic Analysis and Invasive Oxygen Measurements. Mol Imaging Biol 2018; 19:893-902. [PMID: 28409339 DOI: 10.1007/s11307-017-1083-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE The purpose of this study is to use dynamic [18F]fluoromisonidazole ([18F]FMISO) positron emission tomography (PET) to compare estimates of tumor hypoxic fractions (HFs) derived by tracer kinetic modeling, tissue-to-blood ratios (TBR), and independent oxygen (pO2) measurements. PROCEDURES BALB/c mice with EMT6 subcutaneous tumors were selected for PET imaging and invasive pO2 measurements. Data from 120-min dynamic [18F]FMISO scans were fit to two-compartment irreversible three rate constant (K 1, k 2, k 3) and Patlak models (K i). Tumor HFs were calculated and compared using K i, k 3, TBR, and pO2 values. The clinical impact of each method was evaluated on [18F]FMISO scans for three non-small cell lung cancer (NSCLC) radiotherapy patients. RESULTS HFs defined by TBR (≥1.2, ≥1.3, and ≥1.4) ranged from 2 to 85 % of absolute tumor volume. HFs defined by K i (>0.004 ml min cm-3) and k 3 (>0.008 min-1) varied from 9 to 85 %. HF quantification was highly dependent on metric (TBR, k 3, or K i) and threshold. HFs quantified on human [18F]FMISO scans varied from 38 to 67, 0 to 14, and 0.1 to 27 %, for each patient, respectively, using TBR, k 3, and K i metrics. CONCLUSIONS [18F]FMISO PET imaging metric choice and threshold impacts hypoxia quantification reliability. Our results suggest that tracer kinetic modeling has the potential to improve hypoxia quantification clinically as it may provide a stronger correlation with direct pO2 measurements.
Collapse
Affiliation(s)
- Olivia J Kelada
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT, 06520-8040, USA.,Department of Medical Physics in Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Sara Rockwell
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT, 06520-8040, USA.,Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Ming-Qiang Zheng
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yiyun Huang
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Yanfeng Liu
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT, 06520-8040, USA
| | - Carmen J Booth
- Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Roy H Decker
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT, 06520-8040, USA
| | - Uwe Oelfke
- Department of Medical Physics in Radiation Oncology, German Cancer Research Center, Heidelberg, Germany
| | - Richard E Carson
- Department of Diagnostic Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - David J Carlson
- Department of Therapeutic Radiology, Yale University School of Medicine, P.O. Box 208040, New Haven, CT, 06520-8040, USA.
| |
Collapse
|
45
|
Mohamed AS, Bahig H, Aristophanous M, Blanchard P, Kamal M, Ding Y, Cardenas CE, Brock KK, Lai SY, Hutcheson KA, Phan J, Wang J, Ibbott G, Gabr RE, Narayana PA, Garden AS, Rosenthal DI, Gunn GB, Fuller CD. Prospective in silico study of the feasibility and dosimetric advantages of MRI-guided dose adaptation for human papillomavirus positive oropharyngeal cancer patients compared with standard IMRT. Clin Transl Radiat Oncol 2018; 11:11-18. [PMID: 30014042 PMCID: PMC6019867 DOI: 10.1016/j.ctro.2018.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/17/2018] [Accepted: 04/30/2018] [Indexed: 11/18/2022] Open
Abstract
PURPOSE We aim to determine the feasibility and dosimetric benefits of a novel MRI-guided IMRT dose-adaption strategy for human papillomavirus positive (HPV+) oropharyngeal squamous cell carcinoma (OPC). MATERIALS/METHODS Patients with locally advanced HPV+ OPC underwent pre-treatment and in-treatment MRIs every two weeks using RT immobilization setup. For each patient, two IMRT plans were created (i.e. standard and adaptive). The prescription dose for the standard plans was 2.12 Gy/fx for 33 fractions to the initial PTV. For adaptive plans, a new PTVadaptive was generated based on serial MRIs in case of detectable tumor shrinkage. Prescription dose to PTVadaptive was 2.12 Gy/fx to allow for maximum dose to the residual disease. Any previously involved volumes received minimally a floor dose of 50.16 Gy. Uninvolved elective nodal volumes were prescribed 50.16 Gy in 1.52 Gy/fx. Dosimetric parameters of organs at risk (OARs) were recorded for standard vs. adaptive plans. Normal tissue complication probability (NTCP) for toxicity endpoints was calculated using literature-derived multivariate logistic regression models. RESULTS Five patients were included in this pilot study, 3 men and 2 women. Median age was 58 years (range 45-69). Three tumors originated at the tonsillar fossa and two at the base of tongue. The average dose to 95% of initial PTV volume was 70.7 Gy (SD,0.3) for standard plans vs. 58.5 Gy (SD,2.0) for adaptive plans. The majority of OARs showed decrease in dosimetric parameters using adaptive plans vs. standard plans, particularly swallowing related structures. The average reduction in the probability of developing dysphagia ≥ grade2, feeding tube persistence at 6-month post-treatment and hypothyroidism at 1-year post-treatment was 11%, 4%, and 5%, respectively. The probability of xerostomia at 6-month was only reduced by 1% for adaptive plans vs. standard IMRT. CONCLUSION These in silico results showed that the proposed MRI-guided adaptive approach is technically feasible and advantageous in reducing dose to OARs, especially swallowing musculature.
Collapse
Affiliation(s)
- Abdallah S.R. Mohamed
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
- MD Anderson Cancer Center/UT Health Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Houda Bahig
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, Centre hospitalier de l'Université de Montréal (CHUM), Canada
| | - Michalis Aristophanous
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pierre Blanchard
- Department of Radiation Oncology, Institut de Cancérologie Gustave Roussy, Paris, France
| | - Mona Kamal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, University of Ain Shams, Cairo, Egypt
| | - Yao Ding
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carlos E. Cardenas
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristy K. Brock
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephen Y. Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katherine A. Hutcheson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jack Phan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jihong Wang
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Geoffrey Ibbott
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Refaat E. Gabr
- Department of Diagnostic & Interventional Imaging, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ponnada A. Narayana
- Department of Diagnostic & Interventional Imaging, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Adam S. Garden
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David I. Rosenthal
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - G. Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Clifton D. Fuller
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
46
|
Molecular Imaging-Guided Radiotherapy for the Treatment of Head-and-Neck Squamous Cell Carcinoma: Does it Fulfill the Promises? Semin Radiat Oncol 2018; 28:35-45. [PMID: 29173754 DOI: 10.1016/j.semradonc.2017.08.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
With the routine use of intensity modulated radiation therapy for the treatment of head-and-neck squamous cell carcinoma allowing highly conformed dose distribution, there is an increasing need for refining both the selection and the delineation of gross tumor volumes (GTV). In this framework, molecular imaging with positron emission tomography and magnetic resonance imaging offers the opportunity to improve diagnostic accuracy and to integrate tumor biology mainly related to the assessment of tumor cell density, tumor hypoxia, and tumor proliferation into the treatment planning equation. Such integration, however, requires a deep comprehension of the technical and methodological issues related to image acquisition, reconstruction, and segmentation. Until now, molecular imaging has had a limited value for the selection of nodal GTV, but there are increasing evidences that both FDG positron emission tomography and diffusion-weighted magnetic resonance imaging has a potential value for the delineation of the primary tumor GTV, effecting on dose distribution. With the apprehension of the heterogeneity in tumor biology through molecular imaging, growing evidences have been collected over the years to support the concept of dose escalation/dose redistribution using a planned heterogeneous dose prescription, the so-called "dose painting" approach. Validation trials are ongoing, and in the coming years, one may expect to position the dose painting approach in the armamentarium for the treatment of patients with head-and-neck squamous cell carcinoma.
Collapse
|
47
|
Radiomics in Nuclear Medicine Applied to Radiation Therapy: Methods, Pitfalls, and Challenges. Int J Radiat Oncol Biol Phys 2018; 102:1117-1142. [PMID: 30064704 DOI: 10.1016/j.ijrobp.2018.05.022] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/27/2018] [Accepted: 05/02/2018] [Indexed: 02/06/2023]
Abstract
Radiomics is a recent area of research in precision medicine and is based on the extraction of a large variety of features from medical images. In the field of radiation oncology, comprehensive image analysis is crucial to personalization of treatments. A better characterization of local heterogeneity and the shape of the tumor, depicting individual cancer aggressiveness, could guide dose planning and suggest volumes in which a higher dose is needed for better tumor control. In addition, noninvasive imaging features that could predict treatment outcome from baseline scans could help the radiation oncologist to determine the best treatment strategies and to stratify patients as at low risk or high risk of recurrence. Nuclear medicine molecular imaging reflects information regarding biological processes in the tumor thanks to a wide range of radiotracers. Many studies involving 18F-fluorodeoxyglucose positron emission tomography suggest an added value of radiomics compared with the use of conventional PET metrics such as standardized uptake value for both tumor diagnosis and prediction of recurrence or treatment outcome. However, these promising results should not hide technical difficulties that still currently prevent the approach from being widely studied or clinically used. These difficulties mostly pertain to the variability of the imaging features as a function of the acquisition device and protocol, the robustness of the models with respect to that variability, and the interpretation of the radiomic models. Addressing the impact of the variability in acquisition and reconstruction protocols is needed, as is harmonizing the radiomic feature calculation methods, to ensure the reproducibility of studies in a multicenter context and their implementation in a clinical workflow. In this review, we explain the potential impact of positron emission tomography radiomics for radiation therapy and underline the various aspects that need to be carefully addressed to make the most of this promising approach.
Collapse
|
48
|
Gkika E, Oehlke O, Bunea H, Wiedenmann N, Adebahr S, Nestle U, Zamboglou C, Kirste S, Fennell J, Brunner T, Gainey M, Baltas D, Langer M, Urbach H, Bock M, Meyer PT, Grosu AL. Biological imaging for individualized therapy in radiation oncology: part II medical and clinical aspects. Future Oncol 2018. [DOI: 10.2217/fon-2017-0465] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Positron emission tomography and multiparametric MRI provide crucial information concerning tumor extent and normal tissue anatomy. Moreover, they are able to visualize biological characteristics of the tumor, which can be considered in the radiation treatment planning and monitoring. In this review we discuss the impact of biological imaging positron emission tomography and multiparametric MRI for radiation oncology, based on the data of the literature and on the experience of our own institution in this field.
Collapse
Affiliation(s)
- Eleni Gkika
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Oliver Oehlke
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Hatice Bunea
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Sonja Adebahr
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Simon Kirste
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Jamina Fennell
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Thomas Brunner
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Mark Gainey
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Mathias Langer
- Department of Radiology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Horst Urbach
- Department of Neuroradiology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Michael Bock
- Department of Radiology – Medical Physics, Department of Radiology, Faculty of Medicine, Medical Center, University of Freiburg, D-79106, Germany
| | - Philipp T Meyer
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
- Department of Nuclear Medicine, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, D-79106, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| |
Collapse
|
49
|
Salem A, Asselin MC, Reymen B, Jackson A, Lambin P, West CML, O'Connor JPB, Faivre-Finn C. Targeting Hypoxia to Improve Non-Small Cell Lung Cancer Outcome. J Natl Cancer Inst 2018; 110:4096546. [PMID: 28922791 DOI: 10.1093/jnci/djx160] [Citation(s) in RCA: 167] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
Oxygen deprivation (hypoxia) in non-small cell lung cancer (NSCLC) is an important factor in treatment resistance and poor survival. Hypoxia is an attractive therapeutic target, particularly in the context of radiotherapy, which is delivered to more than half of NSCLC patients. However, NSCLC hypoxia-targeted therapy trials have not yet translated into patient benefit. Recently, early termination of promising evofosfamide and tarloxotinib bromide studies due to futility highlighted the need for a paradigm shift in our approach to avoid disappointments in future trials. Radiotherapy dose painting strategies based on hypoxia imaging require careful refinement prior to clinical investigation. This review will summarize the role of hypoxia, highlight the potential of hypoxia as a therapeutic target, and outline past and ongoing hypoxia-targeted therapy trials in NSCLC. Evidence supporting radiotherapy dose painting based on hypoxia imaging will be critically appraised. Carefully selected hypoxia biomarkers suitable for integration within future NSCLC hypoxia-targeted therapy trials will be examined. Research gaps will be identified to guide future investigation. Although this review will focus on NSCLC hypoxia, more general discussions (eg, obstacles of hypoxia biomarker research and developing a framework for future hypoxia trials) are applicable to other tumor sites.
Collapse
Affiliation(s)
- Ahmed Salem
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Marie-Claude Asselin
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Bart Reymen
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Alan Jackson
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Philippe Lambin
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Catharine M L West
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - James P B O'Connor
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Corinne Faivre-Finn
- Division of Cancer Sciences and Division of Informatics, Imaging and Data Sciences, University of Manchester, Manchester, UK; Department of Radiation Oncology (MAASTRO Lab), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| |
Collapse
|
50
|
Mönnich D, Thorwarth D, Leibfarth S, Pfannenberg C, Reischl G, Mauz PS, Nikolaou K, la Fougère C, Zips D, Welz S. Overlap of highly FDG-avid and FMISO hypoxic tumor subvolumes in patients with head and neck cancer. Acta Oncol 2017; 56:1577-1582. [PMID: 28849721 DOI: 10.1080/0284186x.2017.1363910] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND PET imaging may be used to personalize radiotherapy (RT) by identifying radioresistant tumor subvolumes for RT dose escalation. Using the tracers [18F]-fluorodeoxyglucose (FDG) and [18F]-fluoromisonidazole (FMISO), different aspects of tumor biology can be visualized. FDG depicts various biological aspects, e.g., proliferation, glycolysis and hypoxia, while FMISO is more hypoxia specific. In this study, we analyzed size and overlap of volumes based on the two markers for head-and-neck cancer patients (HNSCC). MATERIAL AND METHODS Twenty five HNSCC patients underwent a CT scan, as well as FDG and dynamic FMISO PET/CT prior to definitive radio-chemotherapy in a prospective FMISO dose escalation study. Three PET-based subvolumes of the primary tumor (GTVprim) were segmented: a highly FDG-avid volume VFDG, a hypoxic volume on the static FMISO image acquired four hours post tracer injection (VH) and a retention/perfusion volume (VM) using pharmacokinetic modeling of dynamic FMISO data. Absolute volumes, overlaps and distances to agreement (DTA) were evaluated. RESULTS Sizes of PET-based volumes and the GTVprim are significantly different (GTVprim>VFDG>VH >VM; p < .05). VH is covered by VFDG or DTAs are small (mean coverage 74.4%, mean DTA 1.4 mm). Coverage of VM is less pronounced. With respect to VFDG and VH, the mean coverage is 48.7% and 43.1% and the mean DTA is 5.3 mm and 6.3 mm, respectively. For two patients, DTAs were larger than 2 cm. CONCLUSIONS Hypoxic subvolumes from static PET imaging are typically covered by or in close proximity to highly FDG-avid subvolumes. Therefore, dose escalation to FDG positive subvolumes should cover the static hypoxic subvolumes in most patients, with the disadvantage of larger volumes, resulting in a higher risk of dose-limiting toxicity. Coverage of subvolumes from dynamic FMISO PET is less pronounced. Further studies are needed to explore the relevance of mismatches in functional imaging.
Collapse
Affiliation(s)
- David Mönnich
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Leibfarth
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christina Pfannenberg
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Department of Diagnostic and Interventional Radiology, Preclinical Imaging and Radiopharmacy, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Paul-Stefan Mauz
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Department of Radiology, Nuclear Medicine, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Stefan Welz
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|