51
|
Tumor Voxel Dose-Response Matrix and Dose Prescription Function Derived Using 18F-FDG PET/CT Images for Adaptive Dose Painting by Number. Int J Radiat Oncol Biol Phys 2019; 104:207-218. [PMID: 30684661 DOI: 10.1016/j.ijrobp.2019.01.077] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 01/27/2023]
Abstract
PURPOSE To construct a tumor voxel dose response matrix (DRM) and dose prescription function (DPF) for adaptive dose painting by number (DPbN) based on treatment feedback of fluoro-2-deoxyglucose (FGD) positron emission tomography (PET)/computed tomography (CT) imaging. METHODS AND MATERIALS FDG-PET/CT images obtained before and after chemoradiation therapy and at weekly chemoradiation therapy sessions for each of 18 patients with head and neck cancer, as well as the treatment outcomes, were used in the modeling. All weekly and posttreatment PET/CT images were registered voxel-to-voxel to the corresponding pretreatment baseline PET/CT image. Tumor voxel DRM was created using serial FDG-PET imaging of each patient with respect to the baseline standardized uptake value (SUV0). A tumor voxel control probability (TVCP) lookup table was created using the maximum likelihood estimation on the tumor voxel (SUV0, DRM) domain of all tumors. Tumor voxel DPF was created from the TVCP lookup table and used as the objective function for DPbN-based inverse planning optimization. RESULTS Large intertumoral and intratumoral variations on both tumor voxels (SUV0, DRM) were identified. Tumor voxel dose resistance did not show correlation with its baseline SUV0 value and was the major cause of the tumor local failures. Tumor voxel DPF as the function of tumor voxel (SUV0, DRM) values also showed a very large intertumoral and intratumoral heterogeneity. Most human papillomavirus-negative tumors require a treatment dose >100 Gy to certain local tumor regions. These treatment doses, which are most unlikely to be implementable in conventional radiation therapy, can be achieved using adaptive DPbN treatment. Clinical feasibility was evaluated by comparing the adaptive DPbN treatment plan with the conventional intensity modulated radiation therapy plan. CONCLUSIONS Tumor voxel (SUV0, DRM) provides an intratumoral prognostic map to target tumor locoregional-resistant regions. The corresponding TVCP or DPF provides a quantitative objective to optimize the intratumoral dose distribution for the individuals. The adaptive DPbN with FDG-PET/CT imaging feedback is feasible to implement in clinics.
Collapse
|
52
|
Clinical and Pre-clinical Methods for Quantifying Tumor Hypoxia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1136:19-41. [PMID: 31201714 DOI: 10.1007/978-3-030-12734-3_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hypoxia, a prevalent characteristic of most solid malignant tumors, contributes to diminished therapeutic responses and more aggressive phenotypes. The term hypoxia has two definitions. One definition would be a physiologic state where the oxygen partial pressure is below the normal physiologic range. For most normal tissues, the normal physiologic range is between 10 and 20 mmHg. Hypoxic regions develop when there is an imbalance between oxygen supply and demand. The impact of hypoxia on cancer therapeutics is significant: hypoxic tissue is 3× less radiosensitive than normoxic tissue, the impaired blood flow found in hypoxic tumor regions influences chemotherapy delivery, and the immune system is dependent on oxygen for functionality. Despite the clinical implications of hypoxia, there is not a universal, ideal method for quantifying hypoxia, particularly cycling hypoxia because of its complexity and heterogeneity across tumor types and individuals. Most standard imaging techniques can be modified and applied to measuring hypoxia and quantifying its effects; however, the benefits and challenges of each imaging modality makes imaging hypoxia case-dependent. In this chapter, a comprehensive overview of the preclinical and clinical methods for quantifying hypoxia is presented along with the advantages and disadvantages of each.
Collapse
|
53
|
Skjøtskift T, Evensen ME, Furre T, Moan JM, Amdal CD, Bogsrud TV, Malinen E, Dale E. Dose painting for re-irradiation of head and neck cancer. Acta Oncol 2018; 57:1693-1699. [PMID: 30280623 DOI: 10.1080/0284186x.2018.1512753] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
BACKGROUND For patients with recurrent or second primary disease, re-irradiation can be challenging due to overlap with previously irradiated volumes. Dose painting may be attractive for these patients, as the focus is on delivering maximal dose to areas of high tumor activity. Here, we compare dose painting by contours (DPBC) treatment plans based on 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) with conventional plans. MATERIAL AND METHODS We included 10 patients with recurrent or second primary head and neck cancer (HNC) eligible for re-irradiation. Our conventional re-irradiation regimen is hyperfractionated radiotherapy 1.5 Gy twice daily over 4 weeks, giving a total dose of 60 Gy. For DPBC, we defined two prescription volumes, PV33 and PV66, corresponding to 33 and 66% of the highest FDG uptake in the tumor. The clinical target volume (CTV) prescription dose was 60 Gy, PV33; 65-67 Gy and PV66; 70-73 Gy. The DPBC plan is to be given the first 20 fractions and the conventional plan the last 20 fractions. Dose to organs at risk (OARs) were compared for DPBC and conventional treatment. By summation of the initial curative plan and the re-irradiation plan, we also evaluated differences in dose to the 2 ccm hot spot (D2cc). RESULTS We achieved DPBC plans with adequate target coverage for all 10 patients. There were no significant differences in OAR doses between the standard plans and the DPBC plans (p=.7). Summation of the initial curative plan and the re-irradiation plan showed that the median D2cc increased from 130 Gy (range 113-132 Gy; conventional) to 140 Gy (range 115-145 Gy; DPBC). CONCLUSIONS Our proposed DPBC could be straightforwardly implemented and all plans met the objectives. Re-irradiation of HNC with DPBC may increase tumor control without more side effects compared to conventional radiotherapy.
Collapse
Affiliation(s)
| | | | - Torbjørn Furre
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Jon M. Moan
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | | | - Trond V. Bogsrud
- Department of Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Einar Dale
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
54
|
Schuemann J, Bassler N, Inaniwa T. Computational models and tools. Med Phys 2018; 45:e1073-e1085. [PMID: 30421814 DOI: 10.1002/mp.12521] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 06/21/2017] [Accepted: 08/01/2017] [Indexed: 12/12/2022] Open
Abstract
In this chapter, we describe two different methods, analytical (pencil beam) algorithms and Monte Carlo simulations, used to obtain the intended dose distributions in patients and evaluate their strengths and shortcomings. We discuss the difference between the prescribed physical dose and the biologically effective dose, the relative biological effectiveness (RBE) between ions and photons and the dependence of RBE on the linear energy transfer (LET). Lastly, we show how LET- or RBE-based optimization can be used to improve treatment plans and explore how the availability of multimodality ion beam facilities can be used to design a tumor-specific optimal treatment.
Collapse
Affiliation(s)
- Jan Schuemann
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Niels Bassler
- Medical Radiation Physics, Dept. of Physics, Stockholm University, Sweden
| | - Taku Inaniwa
- Department of Accelerator and Medical Physics, National Institute of Radiological Sciences, QST, Chiba, Japan
| |
Collapse
|
55
|
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: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
56
|
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: 48] [Impact Index Per Article: 6.9] [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
|
57
|
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: 3.9] [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
|
58
|
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: 45] [Impact Index Per Article: 6.4] [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
|
59
|
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: 33] [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
|
60
|
A secondary analysis of FDG spatio-temporal consistency in the randomized phase II PET-boost trial in stage II–III NSCLC. Radiother Oncol 2018; 127:259-266. [DOI: 10.1016/j.radonc.2018.03.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 03/14/2018] [Accepted: 03/15/2018] [Indexed: 12/25/2022]
|
61
|
Gago-Arias A, Sánchez-Nieto B, Espinoza I, Karger CP, Pardo-Montero J. Impact of different biologically-adapted radiotherapy strategies on tumor control evaluated with a tumor response model. PLoS One 2018; 13:e0196310. [PMID: 29698534 PMCID: PMC5919644 DOI: 10.1371/journal.pone.0196310] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 04/10/2018] [Indexed: 11/26/2022] Open
Abstract
Motivated by the capabilities of modern radiotherapy techniques and by the recent developments of functional imaging techniques, dose painting by numbers (DPBN) was proposed to treat tumors with heterogeneous biological characteristics. This work studies different DPBN optimization techniques for virtual head and neck tumors assessing tumor response in terms of cell survival and tumor control probability with a previously published tumor response model (TRM). Uniform doses of 2 Gy are redistributed according to the microscopic oxygen distribution and the density distribution of tumor cells in four virtual tumors with different biological characteristics. In addition, two different optimization objective functions are investigated, which: i) minimize tumor cell survival (OFsurv) or; ii) maximize the homogeneity of the density of surviving tumor cells (OFstd). Several adaptive schemes, ranging from single to daily dose optimization, are studied and the treatment response is compared to that of the uniform dose. The results show that the benefit of DPBN treatments depends on the tumor reoxygenation capability, which strongly differed among the set of virtual tumors investigated. The difference between daily (fraction by fraction) and three weekly optimizations (at the beginning of weeks 1, 3 and 4) was found to be small, and higher benefit was observed for the treatments optimized using OFsurv. This in silico study corroborates the hypothesis that DPBN may be beneficial for treatments of tumors which show reoxygenation during treatment, and that a few optimizations may be sufficient to achieve this therapeutic benefit.
Collapse
Affiliation(s)
- Araceli Gago-Arias
- Instituto de Física, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| | | | - Ignacio Espinoza
- Instituto de Física, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian P. Karger
- National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany
| | - Juan Pardo-Montero
- Grupo de Imaxe Molecular, Instituto de Investigación Sanitaria (IDIS), Santiago de Compostela, Spain
- Servizo de Radiofísica e Protección Radiolóxica, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
62
|
Abstract
Carbon ion therapy is a promising evolving modality in radiotherapy to treat tumors that are radioresistant against photon treatments. As carbon ions are more effective in normal and tumor tissue, the relative biological effectiveness (RBE) has to be calculated by bio-mathematical models and has to be considered in the dose prescription. This review (i) introduces the concept of the RBE and its most important determinants, (ii) describes the physical and biological causes of the increased RBE for carbon ions, (iii) summarizes available RBE measurements in vitro and in vivo, and (iv) describes the concepts of the clinically applied RBE models (mixed beam model, local effect model, and microdosimetric-kinetic model), and (v) the way they are introduced into clinical application as well as (vi) their status of experimental and clinical validation, and finally (vii) summarizes the current status of the use of the RBE concept in carbon ion therapy and points out clinically relevant conclusions as well as open questions. The RBE concept has proven to be a valuable concept for dose prescription in carbon ion radiotherapy, however, different centers use different RBE models and therefore care has to be taken when transferring results from one center to another. Experimental studies significantly improve the understanding of the dependencies and limitations of RBE models in clinical application. For the future, further studies investigating quantitatively the differential effects between normal tissues and tumors are needed accompanied by clinical studies on effectiveness and toxicity.
Collapse
Affiliation(s)
- Christian P Karger
- Department of Medical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany. National Center for Radiation Research in Oncology (NCRO), Heidelberg Institute for Radiation Oncology (HIRO), Heidelberg, Germany. Author to whom any correspondence should be addressed
| | | |
Collapse
|
63
|
Zhu T, Das S, Wong TZ. Integration of PET/MR Hybrid Imaging into Radiation Therapy Treatment. Magn Reson Imaging Clin N Am 2017; 25:377-430. [PMID: 28390536 DOI: 10.1016/j.mric.2017.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Hybrid PET/MR imaging is in early development for treatment planning. This article briefly reviews research and clinical applications of PET/MR imaging in radiation oncology. With improvements in workflow, more specific tracers, and fast and robust acquisition protocols, PET/MR imaging will play an increasingly important role in better target delineation for treatment planning and have clear advantages in the evaluation of tumor response and in a better understanding of tumor heterogeneity. With advances in treatment delivery and the potential of integrating PET/MR imaging with research on radiomics for radiation oncology, quantitative and physiologic information could lead to more precise and personalized RT.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA
| | - Shiva Das
- Department of Radiation Oncology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA
| | - Terence Z Wong
- Department of Radiology, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA.
| |
Collapse
|
64
|
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: 19] [Impact Index Per Article: 2.4] [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
|
65
|
Boeke S, Thorwarth D, Mönnich D, Pfannenberg C, Reischl G, La Fougère C, Nikolaou K, Mauz PS, Paulsen F, Zips D, Welz S. Geometric analysis of loco-regional recurrences in relation to pre-treatment hypoxia in patients with head and neck cancer. Acta Oncol 2017; 56:1571-1576. [PMID: 28891398 DOI: 10.1080/0284186x.2017.1372626] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION A previous pattern-of-failure study has suggested that up to 50% of the loco-regional failures (LRF) in head and neck squamous cell carcinoma (HNSCC) occur outside the initial hypoxic volume determined by [18F]-fluoromisonidazole-PET ([18F]-FMISO-PET). The aim of the present analysis was to correlate spatial patterns of failure with respect to the pretherapeutic dynamic [18F]-FMISO-PET/CT in HNSCC after radiochemotherapy (RCT). MATERIAL AND METHODS Within a running phase 2 trial using [18F]-FMISO-PET imaging prior to RCT in HNSCC patients (n = 54), we have observed so far 11 LRF with a minimum follow-up of 12 months. For nine patients, LRF imaging (CT or [18F]-FDG-PET/CT) for pattern-of-failure analysis was available. Analysis included the static 4-h hypoxic subvolume (VH) as well as a M-parameter volume (VM), which is derived from modeling of dynamic PET. Deformable image registration of the CT scan with the recurrent tumor to the pre-treatment [18F]-FMISO-PET/CT and the planning CT was done to quantify the hypoxic subvolumes compared to the recurrent tumor volume. Moreover, a point-of-origin analysis was performed. RESULTS A total of five local, two regional and two loco-regional recurrences were detected. After deformable image registration of the CT scan with the recurrent tumor to the pre-treatment [18F]-FMISO-PET/CT and the planning CT, a significant overlap of the recurrence volume with [18F]-FMISO-positive subvolumes in the initial gross tumor volume (GTV) was observed. Median overlap of 40.2%, range 9.4-100.0%, for VH and 49.0%, range 4.4-96.4%, for VM was calculated. The point-of-origin analysis showed median distances of 0.0 mm, range 0.0-11.3 mm to VH and 8.6 mm, range 0.0-15.5 mm to VM, respectively. CONCLUSIONS Our data suggest that loco-regional recurrences after RCT originate from the initial GTV (primary tumor and/or lymph node metastases) containing hypoxic subvolumes, which supports the concept of hypoxia imaging-based dose escalation.
Collapse
Affiliation(s)
- Simon Boeke
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Section for Biomedical Physics, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, 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; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - 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; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christina Pfannenberg
- Department of Diagnostic and Interventional Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, Division of Radiology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Christian La Fougère
- Department of Nuclear Medicine, Division of Radiology, 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
| | - Paul-Stefan Mauz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, 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, Tübingen, Germany
| |
Collapse
|
66
|
|
67
|
Shi K, Bayer C, Astner ST, Gaertner FC, Vaupel P, Schwaiger M, Huang SC, Ziegler SI. Quantitative Analysis of [ 18F]FMISO PET for Tumor Hypoxia: Correlation of Modeling Results with Immunohistochemistry. Mol Imaging Biol 2017; 19:120-129. [PMID: 27379986 DOI: 10.1007/s11307-016-0975-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Quantitative evaluation of tumor hypoxia based on H-1-(3-[18F]fluoro-2-hydroxypropyl)-2-nitroimidazole ([18F]FMISO) positron emission tomography (PET) can deliver important information for treatment planning in radiotherapy. However, the merits and limitations of different analysis methods in revealing the underlying physiological feature are not clear. This study aimed to assess these quantitative analysis methods with the support of immunohistological data. PROCEDURES Sixteen nude mice bearing xenografted human squamous cell carcinomas (FaDu or CAL-33) were scanned using 2-h dynamic [18F]FMISO PET. Tumors were resected and sliced, and the hypoxia marker pimonidazole was immunostained followed by H&E staining. The pimonidazole signal was segmented using a k-means clustering algorithm, and the hypoxic fraction (HF) was calculated as the hypoxic area/viable tumor-tissue-area ratio pooled over three tissue slices from the apical, center, and basal layers. PET images were analyzed using various methods including static analysis [standard uptake value (SUV), tumor-to-blood ratio (T/B), tumor-to-muscle ratio (T/M)] and kinetic modeling (Casciari αk A , irreversible and reversible two-tissue compartment k 3, Thorwarth w A k 3, Patlak K i , Logan V d , Cho K), and correlated with HF. RESULTS No significant correlation was found for static analysis. A significant correlation between k 3 of the irreversible two-tissue compartment model and HF was observed (r = 0.61, p = 0.01). The correlation between HF and αk A of the Casciari model could be improved through reducing local minima by testing more sets of initial values (r = 0.59, p = 0.02) or by reducing the model complexity by fixing three parameters (r = 0.63, p = 0.0008). CONCLUSIONS With support of immunohistochemistry data, this study shows that various analysis methods for [18F]FMISO PET perform differently for assessment of tumor hypoxia. A better fitting quality does not necessarily mean a higher physiological correlation. Hypoxia PET analysis needs to consider both the mathematical stability and physiological fidelity. Based on the results of this study, preference should be given to the irreversible two-tissue compartment model as well as the Casciari model with reduced parameters.
Collapse
Affiliation(s)
- Kuangyu Shi
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse. 22, 81675, Munich, Germany.
| | - Christine Bayer
- Department of Radiooncology and Radiotherapy, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Sabrina T Astner
- Department of Radiooncology and Radiotherapy, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Florian C Gaertner
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse. 22, 81675, Munich, Germany
| | - Peter Vaupel
- Department of Radiooncology and Radiotherapy, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse. 22, 81675, Munich, Germany
| | - Sung-Cheng Huang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Sibylle I Ziegler
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaningerstrasse. 22, 81675, Munich, Germany
| |
Collapse
|
68
|
Thorwarth D, Wack LJ, Mönnich D. Hypoxia PET imaging techniques: data acquisition and analysis. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0250-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
69
|
Troost EGC, Koi L, Yaromina A, Krause M. Therapeutic options to overcome tumor hypoxia in radiation oncology. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0247-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
70
|
McMahon SJ, McNamara AL, Schuemann J, Paganetti H, Prise KM. A general mechanistic model enables predictions of the biological effectiveness of different qualities of radiation. Sci Rep 2017; 7:10790. [PMID: 28883414 PMCID: PMC5589818 DOI: 10.1038/s41598-017-10820-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/15/2017] [Indexed: 12/04/2022] Open
Abstract
Predicting the responses of biological systems to ionising radiation is extremely challenging, particularly when comparing X-rays and heavy charged particles, due to the uncertainty in their Relative Biological Effectiveness (RBE). Here we assess the power of a novel mechanistic model of DNA damage repair to predict the sensitivity of cells to X-ray, proton or carbon ion exposures in vitro against over 800 published experiments. By specifying the phenotypic characteristics of cells, the model was able to effectively stratify X-ray radiosensitivity (R2 = 0.74) without the use of any cell-specific fitting parameters. This model was extended to charged particle exposures by integrating Monte Carlo calculated dose distributions, and successfully fit to cellular proton radiosensitivity using a single dose-related parameter (R2 = 0.66). Using these parameters, the model was also shown to be predictive of carbon ion RBE (R2 = 0.77). This model can effectively predict cellular sensitivity to a range of radiations, and has the potential to support developments of personalised radiotherapy independent of radiation type.
Collapse
Affiliation(s)
- Stephen J McMahon
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland. .,Department of Radiation Oncology, Massachusetts General Hospital, 30 Fruit St, Boston, MA, 02114, USA.
| | - Aimee L McNamara
- Department of Radiation Oncology, Massachusetts General Hospital, 30 Fruit St, Boston, MA, 02114, USA
| | - Jan Schuemann
- Department of Radiation Oncology, Massachusetts General Hospital, 30 Fruit St, Boston, MA, 02114, USA
| | - Harald Paganetti
- Department of Radiation Oncology, Massachusetts General Hospital, 30 Fruit St, Boston, MA, 02114, USA
| | - Kevin M Prise
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, BT9 7AE, Northern Ireland
| |
Collapse
|
71
|
Di Perri D, Lee JA, Bol A, Hanin FX, Janssens G, Labar D, Robert A, Sterpin E, Geets X. Correlation analysis of [ 18F]fluorodeoxyglucose and [ 18F]fluoroazomycin arabinoside uptake distributions in lung tumours during radiation therapy. Acta Oncol 2017; 56:1181-1188. [PMID: 28537761 DOI: 10.1080/0284186x.2017.1329594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND PET-guided dose painting (DP) aims to target radioresistant tumour regions in order to improve radiotherapy (RT) outcome. Besides the well-known [18F]fluorodeoxyglucose (FDG), the hypoxia positron emission tomography (PET) tracer [18F]fluoroazomycin arabinoside (FAZA) could provide further useful information to guide the radiation dose prescription. In this study, we compare the spatial distributions of FDG and FAZA PET uptakes in lung tumours. MATERIAL AND METHODS Fourteen patients with unresectable lung cancer underwent FDG and FAZA 4D-PET/CT on consecutive days at three time-points: prior to RT (pre), and during the second (w2), and the third (w3) weeks of RT. All PET/CT were reconstructed in their time-averaged midposition (MidP). The metabolic tumour volume (MTV: FDG standardised uptake value (SUV) > 50% SUVmax), and the hypoxic volume (HV: FAZA SUV > 1.4) were delineated within the gross tumour volume (GTVCT). FDG and FAZA intratumoral PET uptake distributions were subsequently pairwise compared, using both volume-, and voxel-based analyses. RESULTS Volume-based analysis showed large overlap between MTV and HV: median overlapping fraction was 0.90, 0.94 and 0.94, at the pre, w2 and w3 time-points, respectively. Voxel-wise analysis between FDG and FAZA intratumoral PET uptake distributions showed high correlation: median Spearman's rank correlation coefficient was 0.76, 0.77 and 0.76, at the pre, w2 and w3 time-points, respectively. Interestingly, tumours with high FAZA uptake tended to show more similarity between FDG and FAZA intratumoral uptake distributions than those with low FAZA uptake. CONCLUSIONS In unresectable lung carcinomas, FDG and FAZA PET uptake distributions displayed unexpectedly strong similarity, despite the distinct pathways targeted by these tracers. Hypoxia PET with FAZA brought very little added value over FDG from the perspective of DP in this population.
Collapse
Affiliation(s)
- Dario Di Perri
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
- Department of Radiation Oncology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - John A. Lee
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Anne Bol
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - François-Xavier Hanin
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
- Department of Nuclear Medicine, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Daniel Labar
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Annie Robert
- Pole of Epidemiology and Biostatistics (EPID), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Edmond Sterpin
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Xavier Geets
- Center of Molecular Imaging, Radiotherapy and Oncology (MIRO), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain, Brussels, Belgium
- Department of Radiation Oncology, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
72
|
Thorwarth D. Biologically adapted radiation therapy. Z Med Phys 2017; 28:177-183. [PMID: 28869163 DOI: 10.1016/j.zemedi.2017.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 01/05/2023]
Abstract
The aim of biologically adapted radiotherapy (RT) is to shape or paint the prescribed radiation dose according to biological properties of the tumor in order to increase local control rates in the future. Human tumors are known to present with an extremely heterogeneous tissue architecture leading to highly variable local cell densities and chaotic vascular structures leading to tumor hypoxia and regions of increased radiation resistance. The goal of biologically adapted RT or dose painting is to individually adapt the radiation dose to biological features of the tumor as non-invasively assessed with functional imaging in order to overcome increased radiation resistance. This article discusses the whole development chain of biologically adapted RT from radio-biologically relevant processes, functional imaging techniques to visualize tumor biology non-invasively and radiation prescription functions to the implementation of biologically adapted RT in clinical practice.
Collapse
Affiliation(s)
- Daniela Thorwarth
- Sektion Biomedizinische Physik, Universitätsklinikum für Radioonkologie, Eberhard Karls Universität Tübingen, Germany.
| |
Collapse
|
73
|
Löck S, Perrin R, Seidlitz A, Bandurska-Luque A, Zschaeck S, Zöphel K, Krause M, Steinbach J, Kotzerke J, Zips D, Troost EGC, Baumann M. Residual tumour hypoxia in head-and-neck cancer patients undergoing primary radiochemotherapy, final results of a prospective trial on repeat FMISO-PET imaging. Radiother Oncol 2017; 124:533-540. [PMID: 28843726 DOI: 10.1016/j.radonc.2017.08.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND Hypoxia is a well recognised parameter of tumour resistance to radiotherapy, a number of anticancer drugs and potentially immunotherapy. In a previously published exploration cohort of 25 head and neck squamous cell carcinoma (HNSCC) patients on [18F]fluoromisonidazole positron emission tomography (FMISO-PET) we identified residual tumour hypoxia during radiochemotherapy, not before start of treatment, as the driving mechanism of hypoxia-mediated therapy resistance. Several quantitative FMISO-PET parameters were identified as potential prognostic biomarkers. Here we present the results of the prospective validation cohort, and the overall results of the study. METHODS FMISO-PET/CT images of further 25 HNSCC patients were acquired at four time-points before and during radiochemotherapy (RCHT). Peak standardised uptake value, tumour-to-background ratio, and hypoxic volume were analysed. The impact of the potential prognostic parameters on loco-regional tumour control (LRC) was validated by the concordance index (ci) using univariable and multivariable Cox models based on the exploration cohort. Log-rank tests were employed to compare the endpoint between risk groups. RESULTS The two cohorts differed significantly in several baseline parameters, e.g., tumour volume, hypoxic volume, HPV status, and intercurrent death. Validation was successful for several FMISO-PET parameters and showed the highest performance (ci=0.77-0.81) after weeks 1 and 2 of treatment. Cut-off values for the FMISO-PET parameters could be validated after week 2 of RCHT. Median values for the residual hypoxic volume, defined as the ratio of the hypoxic volume in week 2 of RCHT and at baseline, stratified patients into groups of significantly different LRC when applied to the respective other cohort. CONCLUSION Our study validates that residual tumour hypoxia during radiochemotherapy is a major driver of therapy resistance of HNSCC, and that hypoxia after the second week of treatment measured by FMISO-PET may serve as biomarker for selection of patients at high risk of loco-regional recurrence after state-of-the art radiochemotherapy.
Collapse
Affiliation(s)
- Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany
| | - Rosalind Perrin
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Center for Proton Therapy, Paul Scherrer Institute, Switzerland
| | - Annekatrin Seidlitz
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Anna Bandurska-Luque
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Sebastian Zschaeck
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
| | - Klaus Zöphel
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany; Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Jörg Steinbach
- National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Germany
| | - Jörg Kotzerke
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Eberhard Karls Universität Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, Germany
| | - Esther G C Troost
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany.
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Biostatistics and Modeling in Radiation Oncology Group, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, Germany; National Center for Tumor Diseases, partner site Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Germany; Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
74
|
Skórska M, Piotrowski T. Personalized radiotherapy treatment planning based on functional imaging. Rep Pract Oncol Radiother 2017; 22:327-330. [DOI: 10.1016/j.rpor.2017.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 04/19/2017] [Indexed: 11/30/2022] Open
|
75
|
Skorska M, Piotrowski T, Ryczkowski A. Comparison of dose distribution for head and neck cancer patients with and without dose painting escalation during radiotherapy realized with tomotherapy unit. Br J Radiol 2017; 90:20170019. [PMID: 28555505 DOI: 10.1259/bjr.20170019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To determine and quantify the percentage dose increase to organs at risk (OARs) with multiple-level dose painting (DP) for patients with head and neck cancer in comparison with standard regimen. METHODS 12 patients who had undergone fluorine-18 fludeoxyglucose (18F-FDG) positron emission tomography (PET)/CT scan were retrospectively enrolled. Two treatment plans-one using DP escalation and one without-were optimized for each patient base on PET/CT data. The following variables were assessed: dose to OARs and target volumes; execution time; equivalent uniform dose; and normal tissue complication probability. RESULTS No statistically significant differences in beam-on time were observed between plans with and without DP. However, significantly higher doses were observed for all DP-escalated plans in the OARs, with only two exceptions: the brain stem and V60Gy for the mandible. Multiple-level DP resulted in dose increases ranging from 3.0% to 12.9%, depending on the OAR. The largest increase was seen for the parotid glands and the smallest for the mandible. Significant differences in the equivalent uniform dose were observed only for the parotid glands and spinal column, where the dose without DP was lower. The normal tissue complication probability for most OARs was very small. CONCLUSION Importantly, even though DP escalation resulted in higher doses to OARs vs conventional treatment planning, these usually did not exceed the dose tolerance levels. However, clinical trials are necessary to confirm the benefits of DP and to guarantee no additional toxicity. Advances in knowledge: Multiple-level DP by numbers resulted in 3.0-12.9% dose increase, depending on the OAR. Our findings may suggest that DP escalation to very high doses is feasible for about 83% of patients without higher toxicity; however, it still should be confirmed on a larger group of patients.
Collapse
Affiliation(s)
- Malgorzata Skorska
- 1 Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland
| | - Tomasz Piotrowski
- 1 Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland.,2 Department of Electroradiology, University of Medical Sciences, Poznan, Poland
| | - Adam Ryczkowski
- 1 Department of Medical Physics, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
76
|
Grimes DR, Warren DR, Warren S. Hypoxia imaging and radiotherapy: bridging the resolution gap. Br J Radiol 2017; 90:20160939. [PMID: 28540739 PMCID: PMC5603947 DOI: 10.1259/bjr.20160939] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Oxygen distribution is a major determinant of treatment success in radiotherapy, with well-oxygenated tumour regions responding by up to a factor of three relative to anoxic volumes. Conversely, tumour hypoxia is associated with treatment resistance and negative prognosis. Tumour oxygenation is highly heterogeneous and difficult to measure directly. The recent advent of functional hypoxia imaging modalities such as fluorine-18 fluoromisonidazole positron emission tomography have shown promise in non-invasively determining regions of low oxygen tension. This raises the prospect of selectively increasing dose to hypoxic subvolumes, a concept known as dose painting. Yet while this is a promising approach, oxygen-mediated radioresistance is inherently a multiscale problem, and there are still a number of substantial challenges that must be overcome if hypoxia dose painting is to be successfully implemented. Current imaging modalities are limited by the physics of such systems to have resolutions in the millimetre regime, whereas oxygen distribution varies over a micron scale, and treatment delivery is typically modulated on a centimetre scale. In this review, we examine the mechanistic basis and implications of the radiobiological oxygen effect, the factors influencing microscopic heterogeneity in tumour oxygenation and the consequent challenges in the interpretation of clinical hypoxia imaging (in particular fluorine-18 fluoromisonidazole positron emission tomography). We also discuss dose-painting approaches and outline challenges that must be addressed to improve this treatment paradigm.
Collapse
Affiliation(s)
- David Robert Grimes
- 1 Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratory, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX37DQ, UK.,2 Centre for Advanced and Interdisciplinary Radiation Research (CAIRR), School of Mathematics and Physics, Queen's University Belfast, UK
| | - Daniel R Warren
- 1 Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratory, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX37DQ, UK
| | - Samantha Warren
- 1 Cancer Research UK/MRC Oxford Institute for Radiation Oncology, Gray Laboratory, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX37DQ, UK.,3 Hall-Edwards Radiotherapy Research Group, Queen Elizabeth Hospital, Birmingham, UK
| |
Collapse
|
77
|
Nohadani O, Roy A. Robust optimization with time-dependent uncertainty in radiation therapy. ACTA ACUST UNITED AC 2017. [DOI: 10.1080/24725579.2017.1296907] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Omid Nohadani
- Department of Industrial Engineering and Management Sciences, Northwestern University, Evanston, IL, USA
| | - Arkajyoti Roy
- Department of Industrial Engineering and Management Sciences, Northwestern University, Evanston, IL, USA
- Department of Applied Statistics and Operations Research, Bowling Green State University, Bowling Green, OH, USA
| |
Collapse
|
78
|
Welz S, Mönnich D, Pfannenberg C, Nikolaou K, Reimold M, La Fougère C, Reischl G, Mauz PS, Paulsen F, Alber M, Belka C, Zips D, Thorwarth D. Prognostic value of dynamic hypoxia PET in head and neck cancer: Results from a planned interim analysis of a randomized phase II hypoxia-image guided dose escalation trial. Radiother Oncol 2017; 124:526-532. [PMID: 28434798 DOI: 10.1016/j.radonc.2017.04.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/27/2017] [Accepted: 04/02/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE To prospectively assess the prognostic value of tumour hypoxia determined by dynamic [18F]Fluoromisonidazole (dynFMISO) PET/CT, and to evaluate both feasibility and toxicity in patients with locally advanced squamous cell carcinomas of the head and neck (LASCCHN) treated with dynFMISO image-guided dose escalation (DE) using dose-painting by contours. PATIENTS AND METHODS We present a planned interim analysis of a randomized phase II trial. N=25 patients with LASCCHN received baseline dynFMISO PET/CT to derive hypoxic volumes (HV). Patients with tumour hypoxia were randomized into standard radiochemotherapy (stdRT) (70Gy/35 fractions) or DE (77Gy/35 fractions) to the HV. Patients with non-hypoxic tumours were treated with stdRT. Loco-regional control (LRC) in hypoxic patients randomized to stdRT was compared to non-hypoxic patients. Feasibility and toxicity were analysed for patients in the DE arm and compared to stdRT. RESULTS With a mean follow-up of 27months, LRC in hypoxic patients receiving stdRT (n=10) was significantly worse compared to the non-hypoxic group (n=5) (2y-LRC 44.4% versus 100%, p=0.048). The respective LRC for the DE group (n=10) was 70.0%. Treatment compliance as well as acute and late toxicity did not show significant differences between the DE and the standard dose arms. CONCLUSION Tumour hypoxia determined by baseline dynFMISO PET/CT is associated with a high risk of local failure in patients with LASCCHN. First data suggest that DE to HV is feasible without excess toxicity.
Collapse
Affiliation(s)
- Stefan Welz
- Department of Radiation Oncology, University of Tübingen, Germany
| | - David Mönnich
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University of Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University of Tübingen, Germany
| | - Mathias Reimold
- Department of Nuclear Medicine, University of Tübingen, Germany
| | | | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Germany
| | - Paul-Stefan Mauz
- Department of Otorhinolaryngology, University of Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University of Tübingen, Germany
| | - Markus Alber
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Germany; Department of Radiation Oncology, University of Heidelberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University of Tübingen, Germany; Department of Radiation Oncology, LMU Munich, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
79
|
Simoncic U, Leibfarth S, Welz S, Schwenzer N, Schmidt H, Reischl G, Pfannenberg C, Fougère CL, Nikolaou K, Zips D, Thorwarth D. Comparison of DCE-MRI kinetic parameters and FMISO-PET uptake parameters in head and neck cancer patients. Med Phys 2017; 44:2358-2368. [PMID: 28317128 PMCID: PMC5485084 DOI: 10.1002/mp.12228] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 03/08/2017] [Accepted: 03/12/2017] [Indexed: 11/09/2022] Open
Abstract
Purpose Tumor hypoxia is a major cause of radiation resistance, often present in various solid tumors. Dynamic [18F]‐fluoromisonidazole (FMISO) PET imaging is able to reliably assess tumor hypoxia. Comprehensive characterization of tumor microenvironment through FMISO‐PET and dynamic contrast enhanced (DCE) MR multimodality imaging might be a valuable alternative to the dynamic FMISO‐PET acquisition. The aim of this work was to explore the correlation between the FMISO‐PET and DCE‐MRI kinetic parameters. Methods This study was done on head and neck cancer patients (N = 6), who were imaged dynamically with FMISO‐PET and DCE‐MRI on the same day. Images were registered and analyzed for kinetics on a voxel basis. FMISO‐PET images were analyzed with the two‐tissue compartment three rate‐constant model. Additionally, tumor‐to‐muscle ratio (TMR) maps were evaluated. DCE‐MRI was analyzed with the extended Tofts model. Voxel‐wise Pearson's coefficients were calculated for each patient to assess pairwise parameter correlations. Results Median correlations between FMISO uptake parameters and DCE‐MRI kinetic parameters varied across the parameter pairs in the range from −0.05 to 0.71. The highest median correlation of r = 0.71 was observed for the pair Vb−vp, while the K1−Ktrans median correlation was r = 0.45. Median correlation coefficients for the K1−vp and the Ki−Ktrans pairs were r = 0.42 and r = 0.32, respectively. Correlations between FMISO uptake rate parameter Ki and DCE‐MRI kinetic parameters varied substantially across the patients, whereas correlations between the FMISO and DCE‐MRI vascular parameters were consistently high. Median TMR‐K1 and TMR‐Ktrans correlations were r = 0.52 and r = 0.46, respectively, but varied substantially across the patients. Conclusions Based on this clinical evidence, we can conclude that the vascular fraction parameters obtained through DCE‐MRI kinetic analysis or FMISO kinetic analysis measure the same biological property, while other kinetic parameters are unrelated. These results might be useful in the design of future clinical trials involving FMISO‐PET/DCE‐MR multimodality imaging for the assessment of tumor microenvironment.
Collapse
Affiliation(s)
- Urban Simoncic
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany.,Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia.,Jozef Stefan Institute, Ljubljana, Slovenia
| | - Sara Leibfarth
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Stefan Welz
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Nina Schwenzer
- Diagnostic and Interventional Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Holger Schmidt
- Diagnostic and Interventional Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Preclinical Imaging and Radiopharmacy, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Christina Pfannenberg
- Diagnostic and Interventional Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Christian la Fougère
- Nuclear Medicine, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Konstantin Nikolaou
- Diagnostic and Interventional Radiology, Department of Radiology, University Hospital Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| | - Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
80
|
Chvetsov AV, Rajendran JG, Zeng J, Patel SA, Bowen SR, Kim EY. Theoretical effectiveness of cell survival in fractionated radiotherapy with hypoxia-targeted dose escalation. Med Phys 2017; 44:1975-1982. [PMID: 28236652 DOI: 10.1002/mp.12177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/19/2022] Open
Abstract
PURPOSE The goal of this article is to compute the cell survival during fractionated radiotherapy with non-uniform hypoxia-targeted dose distribution relative to cell survival for a uniform dose distribution with equal integral tumor dose. The analysis is performed for different parameters of radiotherapy with conventional and hypofractionated dose regimens. METHODS Our analysis is done using a parsimonious tumor response model that describes the major components of tumor response to radiotherapy such as radiosensitivity, cell proliferation, and hypoxia using as few variables as possible. Two levels of oxygenated and hypoxic cells with the survival curves described by the linear quadratic (LQ) model are implemented in the model. The model allows for analytical solutions for relative cell survival in a single fraction simulation which can be used for additional validation of our numerical simulations. The relative cell survival was computed for conventional and hypofractionated dose regimens in a model problem with radiobiological parameters for the non-small-cell lung cancer. Sensitivity of cell survival to different parameters of radiotherapy such as the relative volume of hypoxic fraction, boost dose ratio, re-oxygenation rate, and delivery errors was investigated. RESULTS Our computational and analytical results show that relative cell survival for non-uniform and uniform dose distributions is larger than 1.0 during the first few fractions of radiotherapy with conventional fractionation. This indicates that the uniform dose distribution produces a higher cell killing effect for the equal integral dose. This may stem from domination of linear contribution to the cell killing effect seen in the dose range of 1-2 Gy and a steeper slope of survival curve in the oxygenated tumor region. This effect can only happen if the distribution of clonogens is nearly uniform; therefore, after the first few fractions, the non-uniform dose distributions outperform the uniform dose distribution and relative cell survival becomes less than 1.0. However, re-oxygenation and delivery errors can also reduce the effectiveness of hypoxia-targeted non-uniform dose distributions toward the end of treatment. For hypofractionated radiotherapy with fractional dose >3 Gy, the relative cell survival was always below 1.0, which means the non-uniform dose distributions produced higher cell killing effect than the uniform dose distribution during the entire treatment. CONCLUSION The data obtained in this work suggest that non-uniform hypoxia-targeted dose distributions are less effective at cell killing than uniform dose distributions at the beginning of radiotherapy with conventional fractionation. However; non-uniform dose distributions can outperform uniform dose distribution by the end of the treatment if the effects of re-oxygenation and delivery errors are reduced. In hypofractionated radiotherapy, non-uniform hypoxia-targeted dose distributions are more efficient than uniform dose distributions during the entire treatment.
Collapse
Affiliation(s)
- Alexei V Chvetsov
- Department of Radiation Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| | - Joseph G Rajendran
- Department of Radiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| | - Jing Zeng
- Department of Radiation Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| | - Shilpen A Patel
- Department of Radiation Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| | - Stephen R Bowen
- Departments of Radiation Oncology and Radiology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| | - Edward Y Kim
- Department of Radiation Oncology, University of Washington, 1959 NE Pacific Street, Seattle, WA, 98195-6043, USA
| |
Collapse
|
81
|
Grönlund E, Johansson S, Montelius A, Ahnesjö A. Dose painting by numbers based on retrospectively determined recurrence probabilities. Radiother Oncol 2017; 122:236-241. [DOI: 10.1016/j.radonc.2016.09.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 09/01/2016] [Accepted: 09/11/2016] [Indexed: 10/20/2022]
|
82
|
Shi K, Bayer C, Gaertner FC, Astner ST, Wilkens JJ, Nüsslin F, Vaupel P, Ziegler SI. Matching the reaction-diffusion simulation to dynamic [ 18F]FMISO PET measurements in tumors: extension to a flow-limited oxygen-dependent model. Physiol Meas 2017; 38:188-204. [PMID: 28055983 DOI: 10.1088/1361-6579/aa5071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Positron-emission tomography (PET) with hypoxia specific tracers provides a noninvasive method to assess the tumor oxygenation status. Reaction-diffusion models have advantages in revealing the quantitative relation between in vivo imaging and the tumor microenvironment. However, there is no quantitative comparison of the simulation results with the real PET measurements yet. The lack of experimental support hampers further applications of computational simulation models. This study aims to compare the simulation results with a preclinical [18F]FMISO PET study and to optimize the reaction-diffusion model accordingly. Nude mice with xenografted human squamous cell carcinomas (CAL33) were investigated with a 2 h dynamic [18F]FMISO PET followed by immunofluorescence staining using the hypoxia marker pimonidazole and the endothelium marker CD 31. A large data pool of tumor time-activity curves (TAC) was simulated for each mouse by feeding the arterial input function (AIF) extracted from experiments into the model with different configurations of the tumor microenvironment. A measured TAC was considered to match a simulated TAC when the difference metric was below a certain, noise-dependent threshold. As an extension to the well-established Kelly model, a flow-limited oxygen-dependent (FLOD) model was developed to improve the matching between measurements and simulations. The matching rate between the simulated TACs of the Kelly model and the mouse PET data ranged from 0 to 28.1% (on average 9.8%). By modifying the Kelly model to an FLOD model, the matching rate between the simulation and the PET measurements could be improved to 41.2-84.8% (on average 64.4%). Using a simulation data pool and a matching strategy, we were able to compare the simulated temporal course of dynamic PET with in vivo measurements. By modifying the Kelly model to a FLOD model, the computational simulation was able to approach the dynamic [18F]FMISO measurements in the investigated tumors.
Collapse
Affiliation(s)
- Kuangyu Shi
- Department of Nuclear Medicine, Technische Universität München, Klinikum rechts der Isar, Germany
| | | | | | | | | | | | | | | |
Collapse
|
83
|
Abouzied MM, Fathala A, Alsugair A, Muhaideb AIA, Qahtani MHA. Role of Fluorodeoxyglucose-Positron Emission Tomography/Computed Tomography in the Evaluation of Head and Neck Carcinoma. World J Nucl Med 2017; 16:257-265. [PMID: 29033672 PMCID: PMC5639440 DOI: 10.4103/wjnm.wjnm_40_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fluorodeoxyglucose (FDG)-positron emission tomography-computed tomography (PET-CT) has been playing a pivotal role in tumor imaging for the past 20 years. Head and neck (HN) cancers are a good example that can illustrate such unique role of FDG imaging contributing to the patient's management. In this review article, we will describe the normal physiological distribution of FDG within HN structures focusing on its limitations and pitfalls. In addition, we will be also describing its role in the initial staging and restaging of the disease, particularly with regard to therapy response assessment. Furthermore, its role in the evaluation of patients with malignant cervical adenopathy from an unknown primary will be described. In 2016, the Royal College of Radiologists in its third edition published evidence-based guidelines for PET-CT use in HN cancer emphasizing its rule in all these clinical scenarios that are being described in this review. Finally, we will be highlighting future directions in the field of molecular imaging of HN tumors with a special emphasis on the new PET tracers.
Collapse
Affiliation(s)
- Moheieldin M Abouzied
- Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Ahmed Fathala
- Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Abdulaziz Alsugair
- Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Ahmad I Al Muhaideb
- Department of Radiology, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| | - Mohammed H Al Qahtani
- Department of Cyclotron and Radiopharmaceuticals, King Faisal Specialist Hospital and Research Center, Riyadh 11211, Saudi Arabia
| |
Collapse
|
84
|
Warren DR, Partridge M. The role of necrosis, acute hypoxia and chronic hypoxia in 18F-FMISO PET image contrast: a computational modelling study. Phys Med Biol 2016; 61:8596-8624. [PMID: 27880734 PMCID: PMC5717515 DOI: 10.1088/1361-6560/61/24/8596] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 09/14/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
Positron emission tomography (PET) using 18F-fluoromisonidazole (FMISO) is a promising technique for imaging tumour hypoxia, and a potential target for radiotherapy dose-painting. However, the relationship between FMISO uptake and oxygen partial pressure ([Formula: see text]) is yet to be quantified fully. Tissue oxygenation varies over distances much smaller than clinical PET resolution (<100 μm versus ∼4 mm), and cyclic variations in tumour perfusion have been observed on timescales shorter than typical FMISO PET studies (∼20 min versus a few hours). Furthermore, tracer uptake may be decreased in voxels containing some degree of necrosis. This work develops a computational model of FMISO uptake in millimetre-scale tumour regions. Coupled partial differential equations govern the evolution of oxygen and FMISO distributions, and a dynamic vascular source map represents temporal variations in perfusion. Local FMISO binding capacity is modulated by the necrotic fraction. Outputs include spatiotemporal maps of [Formula: see text] and tracer accumulation, enabling calculation of tissue-to-blood ratios (TBRs) and time-activity curves (TACs) as a function of mean tissue oxygenation. The model is characterised using experimental data, finding half-maximal FMISO binding at local [Formula: see text] of 1.4 mmHg (95% CI: 0.3-2.6 mmHg) and half-maximal necrosis at 1.2 mmHg (0.1-4.9 mmHg). Simulations predict a non-linear non-monotonic relationship between FMISO activity (4 hr post-injection) and mean tissue [Formula: see text] : tracer uptake rises sharply from negligible levels in avascular tissue, peaking at ∼5 mmHg and declining towards blood activity in well-oxygenated conditions. Greater temporal variation in perfusion increases peak TBRs (range 2.20-5.27) as a result of smaller predicted necrotic fraction, rather than fundamental differences in FMISO accumulation under acute hypoxia. Identical late FMISO uptake can occur in regions with differing [Formula: see text] and necrotic fraction, but simulated TACs indicate that additional early-phase information may allow discrimination of hypoxic and necrotic signals. We conclude that a robust approach to FMISO interpretation (and dose-painting prescription) is likely to be based on dynamic PET analysis.
Collapse
Affiliation(s)
- Daniel R Warren
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Mike Partridge
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
85
|
Feasibility of voxel-based Dose Painting for recurrent Glioblastoma guided by ADC values of Diffusion-Weighted MR imaging. Phys Med 2016; 32:1651-1658. [DOI: 10.1016/j.ejmp.2016.11.106] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 10/26/2016] [Accepted: 11/15/2016] [Indexed: 01/01/2023] Open
|
86
|
Panek R, Schmidt MA, Borri M, Koh DM, Riddell A, Welsh L, Dunlop A, Powell C, Bhide SA, Nutting CM, Harrington KJ, Newbold KL, Leach MO. Time-resolved angiography with stochastic trajectories for dynamic contrast-enhanced MRI in head and neck cancer: Are pharmacokinetic parameters affected? Med Phys 2016; 43:6024. [PMID: 27806585 DOI: 10.1118/1.4964795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/26/2016] [Accepted: 09/30/2016] [Indexed: 12/31/2022] Open
Abstract
PURPOSE To investigate the effects of different time-resolved angiography with stochastic trajectories (TWIST) k-space undersampling schemes on calculated pharmacokinetic dynamic contrast-enhanced (DCE) vascular parameters. METHODS A digital perfusion phantom was employed to simulate effects of TWIST on characteristics of signal changes in DCE. Furthermore, DCE-MRI was acquired without undersampling in a group of patients with head and neck squamous cell carcinoma and used to simulate a range of TWIST schemes. Errors were calculated as differences between reference and TWIST-simulated DCE parameters. Parametrical error maps were used to display the averaged results from all tumors. RESULTS For a relatively wide range of undersampling schemes, errors in pharmacokinetic parameters due to TWIST were under 10% for the volume transfer constant, Ktrans, and total extracellular extravascular space volume, Ve. TWIST induced errors in the total blood plasma volume, Vp, were the largest observed, and these were inversely dependent on the area of the fully sampled k-space. The magnitudes of errors were not correlated with Ktrans, Vp and weakly correlated with Ve. CONCLUSIONS The authors demonstrated methods to validate and optimize k-space view-sharing techniques for pharmacokinetic DCE studies using a range of clinically relevant spatial and temporal patient derived data. The authors found a range of undersampling patterns for which the TWIST sequence can be reliably used in pharmacokinetic DCE-MRI. The parameter maps created in the study can help to make a decision between temporal and spatial resolution demands and the quality of enhancement curve characterization.
Collapse
Affiliation(s)
- Rafal Panek
- CR-UK Cancer Imaging Centre, London SM2 5PT, United Kingdom; The Institute of Cancer Research, London SM2 5PT, United Kingdom; and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Maria A Schmidt
- CR-UK Cancer Imaging Centre, London SM2 5PT, United Kingdom; The Institute of Cancer Research, London SM2 5PT, United Kingdom; and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Marco Borri
- CR-UK Cancer Imaging Centre, London SM2 5PT, United Kingdom; The Institute of Cancer Research, London SM2 5PT, United Kingdom; and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Dow-Mu Koh
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Angela Riddell
- The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Liam Welsh
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Alex Dunlop
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Ceri Powell
- The Institute of Cancer Research, London SM2 5PT, United Kingdom
| | - Shreerang A Bhide
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Christopher M Nutting
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Kevin J Harrington
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Kate L Newbold
- The Institute of Cancer Research, London SM2 5PT, United Kingdom and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| | - Martin O Leach
- CR-UK Cancer Imaging Centre, London SM2 5PT, United Kingdom; The Institute of Cancer Research, London SM2 5PT, United Kingdom; and The Royal Marsden NHS Trust, London SM2 5PT, United Kingdom
| |
Collapse
|
87
|
Bittner MI, Wiedenmann N, Bucher S, Hentschel M, Mix M, Rücker G, Weber WA, Meyer PT, Werner M, Grosu AL, Kayser G. Analysis of relation between hypoxia PET imaging and tissue-based biomarkers during head and neck radiochemotherapy. Acta Oncol 2016; 55:1299-1304. [PMID: 27593107 DOI: 10.1080/0284186x.2016.1219046] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Tumor hypoxia is associated with poor prognosis and outcome and can be visualized using 18F-MISO-positron emission tomography (PET) imaging. The goal of this study was to evaluate the correlation between biological markers and biological imaging in a group of patients in whom a correlation between biological imaging and outcome has previously been demonstrated. MATERIAL AND METHODS In a prospective pilot project, 16 patients with locally advanced cancer of the head and neck underwent 18F-MISO-PET scans before and during primary radiochemotherapy in addition to 18F-FDG-PET and computed tomography (CT). Tumor biopsies were stained for three tissue-based markers (Ku80, CAIX, CD44); in addition, human papillomavirus (HPV) status was assessed. H-scores of marker expression were generated and the results were correlated with the biological imaging and clinical outcome. RESULTS No statistically significant correlation was established between the H-scores for Ku80, CD44 and CAIX or between any of the H-scores and the imaging variables (tumor volume on 18F-FDG-PET in ml, hypoxic subvolume as assessed by 18F-MISO-PET in ml, and SUVmax tumor/SUVmean muscle during the 18F-MISO-PET). A statistically significant negative correlation was found between CD44 H-score and HPV status (p = .004). Cox regression analysis for overall survival and recurrence-free survival showed one significant result for CAIX being associated with improved overall survival [hazard ratio 0.96 (0.93-1.00), p = .047]. CONCLUSION Expression of Ku80, CAIX and CD44 as assessed by immunohistochemistry of tumor biopsies were not correlated to one another or the biological imaging data. However, there was a significant influence of CAIX on overall survival and between CD44 and HPV.
Collapse
Affiliation(s)
- Martin-Immanuel Bittner
- Department of Radiation Oncology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, UK
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sabine Bucher
- Department of Radiation Oncology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Hentschel
- Department of Radiation Oncology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Inselspital Bern, Bern, Switzerland
| | - Michael Mix
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gerta Rücker
- Institute for Medical Biometry and Statistics, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Wolfgang A. Weber
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Molecular Imaging and Therapy Service, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Philipp T. Meyer
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Martin Werner
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Surgical Pathology, Department of Pathology, Medical Center?University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center–University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Gian Kayser
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Surgical Pathology, Department of Pathology, Medical Center?University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
88
|
Saberian F, Ghate A, Kim M. A theoretical stochastic control framework for adapting radiotherapy to hypoxia. Phys Med Biol 2016; 61:7136-7161. [DOI: 10.1088/0031-9155/61/19/7136] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
89
|
Abstract
CLINICAL/METHODICAL ISSUE The aim of magnetic resonance imaging (MRI) guided radiotherapy is high precision in treatment delivery. With new developments it is possible to focus the high dose irradiation on the tumor while sparing the surrounding tissue. The achievements in precision of the treatment planning and delivery warrant equally precise tumor definition. STANDARD RADIOLOGICAL METHODS In conventional radiation therapy it is necessary to carry out a planning computed tomography (CT). For many tumors there is also need for an additional morphological MRI because of more accurate tumor definition. In standard radiotherapy the tumor volume is irradiated with a homogeneous dose. METHODICAL INNOVATIONS The aim of functional multiparametric MRI is to visualize and quantify biological, physiological and pathological processes at the cellular and molecular levels. Based on this information it is possible to elucidate tumor biology and identify subvolumes of more aggressive behavior. They are often radiotherapy-resistant, leading to tumor recurrence thus requiring further dose escalation. The concept of inhomogeneous tumor irradiation according to its biological behavior is called dose painting. PERFORMANCE Dose painting is technically feasible. The expected clinical benefit is motivated by selective treatment adaptations based on biological tumor characteristics. Tumors show variable response to therapy underlining the need for individual treatment plans. This approach may lead not only to higher local control but also to better sparing of normal surrounding tissue. ACHIEVEMENTS With the clinical implementation of dose painting, improvements in the therapeutic outcome can be expected. PRACTICAL RECOMMENDATIONS Due to the existing technical challenges, extensive collaboration between radiation oncologists, radiologists, medical physicists and radiation biologists is needed.
Collapse
Affiliation(s)
- P Georg
- EBG MedAustron GmbH, Marie-Curie-Straße 5, 2700, Wiener Neustadt, Österreich. .,Christian Doppler Labor für die Medizinische Strahlenforschung, Medizinische Universität Wien, Wien, Österreich.
| | - P Andrzejewski
- Christian Doppler Labor für die Medizinische Strahlenforschung, Medizinische Universität Wien, Wien, Österreich.,Abteilung für medizinische Strahlenphysik, Univ. Klinik für Strahlentherapie, Medizinische Universität Wien, Wien, Österreich
| | - K Pinker
- Christian Doppler Labor für die Medizinische Strahlenforschung, Medizinische Universität Wien, Wien, Österreich.,Abteilung für molekulare Bildgebung, Univ. Klinik für Radiologie und Nuklearmedizin, Medizinische Universität Wien, Wien, Österreich
| | - D Georg
- Christian Doppler Labor für die Medizinische Strahlenforschung, Medizinische Universität Wien, Wien, Österreich.,Abteilung für medizinische Strahlenphysik, Univ. Klinik für Strahlentherapie, Medizinische Universität Wien, Wien, Österreich
| |
Collapse
|
90
|
Mönnich D, Welz S, Thorwarth D, Pfannenberg C, Reischl G, Mauz PS, Nikolaou K, la Fougère C, Zips D. Robustness of quantitative hypoxia PET image analysis for predicting local tumor control. Acta Oncol 2016; 54:1364-9. [PMID: 26481464 DOI: 10.3109/0284186x.2015.1071496] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Previous studies suggested the maximum tumor to background ratio (TBRmax) in FMISO PET images as a potentially predictive parameter for local control after radio-chemotherapy (CRT) in head and neck squamous cell carcinomas (HNSCC). However, different TBRmax thresholds for stratification were reported, implying that a common threshold cannot readily be used among different institutions without the risk of reducing prediction accuracy. Therefore, this study investigated the robustness of using a common pre-defined TBRmax, simulating a multicenter clinical trial. MATERIAL AND METHODS FMISO PET/CT was performed four hours post-injection in 22 patients with advanced HNSCC in a phase II FMISO dose escalation study. PET background regions of interest (ROIs) were manually defined in deep neck muscles. TBRmax was calculated as the mean of the highest-valued voxels within the high risk RT planning target volume. Its predictive power with respect to local control was tested, classifying patients using median TBRmax as threshold. The influence of systematically varying quantification between institutions was studied in silico by applying offsets of ± 10% and ± 20% to the TBRmax of all patients, while the threshold remained constant. The effect was analyzed using a receiver operating characteristic (ROC). True positive and false positive rates (TPR/FPR) as well as positive and negative predictive values (PPV/NPV) were evaluated. RESULTS For the reference condition without an offset the median TBRmax was 2.0 (1.4-3.5). Patients were classified using this threshold and TPR = 0.7, FPR = 0.4, PPV = 0.5 and NPV = 0.8 were observed. Accuracy declined with increasing offsets. Negative offsets of -10% and -20% resulted in TPR = 0.43 and 0.14, FPR = 0.20 and 0.13, PPV = 0.50 and 0.33 and NPV = 0.75 and 0.68, respectively. Positive offsets of + 10% and + 20% resulted in TPR = 1.00 and 1.00, FPR = 0.53 and 0.67, PPV = 0.47 and 0.41 and NPV = 1.00 and 1.00, respectively. CONCLUSIONS Using a common pre-defined TBRmax threshold in multicenter trials requires careful standardization and harmonization of all steps from patient preparation to image analysis. Our results indicate that TBRmax should deviate less than 10% from reference conditions (absolute value in this dataset ± 0.2). This conclusion likely applies to all low contrast nitroimidazole hypoxia PET tracers.
Collapse
Affiliation(s)
- David Mönnich
- a Section for Biomedical Physics, Department of Radiation Oncology , Eberhard Karls University Tübingen , Germany
- b German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Stefan Welz
- c Department of Radiation Oncology , Eberhard Karls University Tübingen , Germany
| | - Daniela Thorwarth
- a Section for Biomedical Physics, Department of Radiation Oncology , Eberhard Karls University Tübingen , Germany
| | - Christina Pfannenberg
- d Department of Diagnostic and Interventional Radiology , Eberhard Karls University Tübingen , Germany
| | - Gerald Reischl
- e Department of Preclinical Imaging and Radiopharmacy , Eberhard Karls University Tübingen , Germany
| | - Paul-Stefan Mauz
- f Department of Otorhinolaryngology , Head and Neck Surgery, Eberhard Karls University Tübingen , Germany
| | - Konstantin Nikolaou
- d Department of Diagnostic and Interventional Radiology , Eberhard Karls University Tübingen , Germany
| | | | - Daniel Zips
- c Department of Radiation Oncology , Eberhard Karls University Tübingen , Germany
| |
Collapse
|
91
|
Wack LJ, Mönnich D, Yaromina A, Zips D, Baumann M, Thorwarth D. Correlation of FMISO simulations with pimonidazole-stained tumor xenografts: A question of O2 consumption? Med Phys 2016; 43:4113. [PMID: 27370131 DOI: 10.1118/1.4951728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE To compare a dedicated simulation model for hypoxia PET against tumor microsections stained for different parameters of the tumor microenvironment. The model can readily be adapted to a variety of conditions, such as different human head and neck squamous cell carcinoma (HNSCC) xenograft tumors. METHODS Nine different HNSCC tumor models were transplanted subcutaneously into nude mice. Tumors were excised and immunoflourescently labeled with pimonidazole, Hoechst 33342, and CD31, providing information on hypoxia, perfusion, and vessel distribution, respectively. Hoechst and CD31 images were used to generate maps of perfused blood vessels on which tissue oxygenation and the accumulation of the hypoxia tracer FMISO were mathematically simulated. The model includes a Michaelis-Menten relation to describe the oxygen consumption inside tissue. The maximum oxygen consumption rate M0 was chosen as the parameter for a tumor-specific optimization as it strongly influences tracer distribution. M0 was optimized on each tumor slice to reach optimum correlations between FMISO concentration 4 h postinjection and pimonidazole staining intensity. RESULTS After optimization, high pixel-based correlations up to R(2) = 0.85 were found for individual tissue sections. Experimental pimonidazole images and FMISO simulations showed good visual agreement, confirming the validity of the approach. Median correlations per tumor model varied significantly (p < 0.05), with R(2) ranging from 0.20 to 0.54. The optimum maximum oxygen consumption rate M0 differed significantly (p < 0.05) between tumor models, ranging from 2.4 to 5.2 mm Hg/s. CONCLUSIONS It is feasible to simulate FMISO distributions that match the pimonidazole retention patterns observed in vivo. Good agreement was obtained for multiple tumor models by optimizing the oxygen consumption rate, M0, whose optimum value differed significantly between tumor models.
Collapse
Affiliation(s)
- L J Wack
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen 72076, Germany
| | - D Mönnich
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen 72076, Germany; German Cancer Consortium (DKTK), Tübingen 72076, Germany; and German Cancer Research Center (DKFZ), Heidelberg 69121, Germany
| | - A Yaromina
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01309, Germany and Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht 6229 ET, The Netherlands
| | - D Zips
- German Cancer Consortium (DKTK), Tübingen 72076, Germany; German Cancer Research Center (DKFZ), Heidelberg 69121, Germany and Department of Radiation Oncology, University Hospital Tübingen, Tübingen 72076, Germany
| | - M Baumann
- German Cancer Consortium (DKTK), Dresden 01307, Germany; German Cancer Research Center (DKFZ), Heidelberg 69121, Germany; OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01309, Germany; Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; and Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden 01328, Germany
| | - D Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, Tübingen 72076, Germany
| |
Collapse
|
92
|
Okamoto S, Shiga T, Yasuda K, Watanabe S, Hirata K, Nishijima KI, Magota K, Kasai K, Onimaru R, Tuchiya K, Kuge Y, Shirato H, Tamaki N. The reoxygenation of hypoxia and the reduction of glucose metabolism in head and neck cancer by fractionated radiotherapy with intensity-modulated radiation therapy. Eur J Nucl Med Mol Imaging 2016; 43:2147-2154. [DOI: 10.1007/s00259-016-3431-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 05/23/2016] [Indexed: 12/19/2022]
|
93
|
Xu S, Wu Z, Yang C, Ma L, Qu B, Chen G, Yao W, Wang S, Liu Y, Li XA. Radiation-induced CT number changes in GTV and parotid glands during the course of radiation therapy for nasopharyngeal cancer. Br J Radiol 2016; 89:20140819. [PMID: 27033059 DOI: 10.1259/bjr.20140819] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To investigate the changes in CT number (CTN) in gross tumour volume (GTV) and organs at risk (OARs) during the course of radiation therapy (RT) for nasopharyngeal cancer (NPC). METHODS Daily megavoltage CT (MVCT) data collected from 30 patients with NPC treated with a prescription dose of 70 Gy in 30-33 fractions using helical tomotherapy were retrospectively analyzed. The contours of GTV and OARs on daily MVCTs were obtained by populating the planning contours from planning CT to daily MVCTs with manual editing, if necessary. The changes of GTV and OAR volumes and the histograms of CTN in the GTV and OARs during the course of RT delivery were analyzed. RESULTS Volumes of GTV and parotid glands were reduced during the course of radiation treatment, with an average shrinkage rate of 0.23% per day (range, 0.02-0.8%) and 1.2% per day (range, 0.2-2.3%), respectively. The mean CTN changes in GTV and ipsilateral and contralateral parotid glands were reduced by 52 ± 35 HU, 18 ± 20 HU and 17 ± 22 HU, respectively. For GTV, the CTN and GTV volume decreases were found to be correlated with each other (p < 0.0001). No noticeable CTN change was found in the spinal cord and non-specified tissue irradiated with low doses. CONCLUSION The CTN changes in GTV and parotids are measurable during the delivery of fractionated radiotherapy for NPC, were associated with the doses received (the number of fractions delivered) and were patient specific. ADVANCES IN KNOWLEDGE The CTN change during radiotherapy is dose dependent and is measurable for NPC.
Collapse
Affiliation(s)
- Shouping Xu
- 1 Key Laboratory of Particle and Radiation Imaging, Tsinghua University, Ministry of Education, Beijing, China.,2 Department of Radiation Oncology, PLA General Hospital, Beijing, China.,3 Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhaoxia Wu
- 1 Key Laboratory of Particle and Radiation Imaging, Tsinghua University, Ministry of Education, Beijing, China
| | - Cungeng Yang
- 3 Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Lin Ma
- 2 Department of Radiation Oncology, PLA General Hospital, Beijing, China
| | - Baolin Qu
- 2 Department of Radiation Oncology, PLA General Hospital, Beijing, China
| | - Guangpei Chen
- 3 Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Weirong Yao
- 2 Department of Radiation Oncology, PLA General Hospital, Beijing, China
| | - Shi Wang
- 1 Key Laboratory of Particle and Radiation Imaging, Tsinghua University, Ministry of Education, Beijing, China
| | - Yaqiang Liu
- 1 Key Laboratory of Particle and Radiation Imaging, Tsinghua University, Ministry of Education, Beijing, China
| | - X Allen Li
- 3 Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
94
|
18F-FDG PET/CT quantification in head and neck squamous cell cancer: principles, technical issues and clinical applications. Eur J Nucl Med Mol Imaging 2016; 43:1360-75. [DOI: 10.1007/s00259-015-3294-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 12/14/2015] [Indexed: 01/28/2023]
|
95
|
Chirla R, Marcu LG. PET-based quantification of statistical properties of hypoxic tumor subvolumes in head and neck cancer. Phys Med 2016; 32:23-35. [DOI: 10.1016/j.ejmp.2015.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 11/29/2015] [Accepted: 12/13/2015] [Indexed: 11/30/2022] Open
|
96
|
Zschaeck S, Steinbach J, Troost EGC. FMISO as a Biomarker for Clinical Radiation Oncology. Recent Results Cancer Res 2016; 198:189-201. [PMID: 27318688 DOI: 10.1007/978-3-662-49651-0_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumour hypoxia is a well-known negative prognostic marker in almost all solid tumours. [18F]Fluoromisonidazole (FMISO)-positron emission tomography (PET) is a non-invasive method to detect tumour hypoxia. Compared to other methods of hypoxia assessment it possesses some considerable advantages: It is non-invasive, it delivers spatial information on the hypoxia distribution within the entire tumour volume, and it can be repeated during the course of radio(chemo)therapy. This chapter briefly describes different methods of hypoxia evaluation and focuses on hypoxia PET imaging, with the most commonly used tracer being FMISO. The preclinical rationale and clinical studies to use FMISO-PET for patient stratification in radiation therapy are discussed as well as possible agents or radiation-dose modifications to overcome hypoxia.
Collapse
Affiliation(s)
- Sebastian Zschaeck
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany. .,German Cancer Consortium (DKTK), Dresden, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Esther G C Troost
- OncoRay - National Center for Radiation Research in Oncology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden and Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,German Cancer Consortium (DKTK), Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstrasse 74, 01307, Dresden, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| |
Collapse
|
97
|
Rajendran JG, Krohn KA. F-18 fluoromisonidazole for imaging tumor hypoxia: imaging the microenvironment for personalized cancer therapy. Semin Nucl Med 2015; 45:151-62. [PMID: 25704387 DOI: 10.1053/j.semnuclmed.2014.10.006] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Hypoxia in solid tumors is one of the seminal mechanisms for developing aggressive trait and treatment resistance in solid tumors. This evolutionarily conserved biological mechanism along with derepression of cellular functions in cancer, although resulting in many challenges, provide us with opportunities to use these adversities to our advantage. Our ability to use molecular imaging to characterize therapeutic targets such as hypoxia and apply this information for therapeutic interventions is growing rapidly. Evaluation of hypoxia and its biological ramifications to effectively plan appropriate therapy that can overcome the cure-limiting effects of hypoxia provides an objective means for treatment selection and planning. Fluoromisonidazole (FMISO) continues to be the lead radiopharmaceutical in PET imaging for the evaluation, prognostication, and quantification of tumor hypoxia, one of the key elements of the tumor microenvironment. FMISO is less confounded by blood flow, and although the images have less contrast than FDG-PET, its uptake after 2 hours is an accurate reflection of inadequate regional oxygen partial pressure at the time of radiopharmaceutical administration. By virtue of extensive clinical utilization, FMISO remains the lead candidate for imaging and quantifying hypoxia. The past decade has seen significant technological advances in investigating hypoxia imaging in radiation treatment planning and in providing us with the ability to individualize radiation delivery and target volume coverage. The presence of widespread hypoxia in the tumor can be effectively targeted with a systemic hypoxic cell cytotoxin or other agents that are more effective with diminished oxygen partial pressure, either alone or in combination. Molecular imaging in general and hypoxia imaging in particular will likely become an important in vivo imaging biomarker of the future, complementing the traditional direct tissue sampling methods by providing a snap shot of a primary tumor and metastatic disease and in following treatment response and will serve as adjuncts to personalized therapy.
Collapse
Affiliation(s)
- Joseph G Rajendran
- Department of Radiology, University of Washington, Seattle, WA; Department of Radiation Oncology, University of Washington, Seattle, WA.
| | - Kenneth A Krohn
- Department of Radiology, University of Washington, Seattle, WA; Department of Radiation Oncology, University of Washington, Seattle, WA
| |
Collapse
|
98
|
Musunuru HB, Loblaw A. Clinical trials of stereotactic ablative radiotherapy for prostate cancer: updates and future direction. Future Oncol 2015; 11:819-31. [PMID: 25757684 DOI: 10.2217/fon.15.14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Stereotactic body radiotherapy, also known as stereotactic ablative body radiotherapy (SABR), is an emerging treatment option for lung, prostate, liver and other tumors. Key factors in SABR are delivery of a high-dose radiation per fraction, proper patient positioning and target localization. Our review details the various radiotherapy techniques, dose fractionation schedules and toxicities for prostate SABR. Ongoing Phase II/III SABR studies across various risk groups have been included. It also discusses the role of conscientious focal dose escalation of the dominant intraprostatic nodule, integrating multiparametric MRI into radiotherapy protocols and finally cost-effectiveness of SABR.
Collapse
Affiliation(s)
- H Bindu Musunuru
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, 2075 Bayview Avenue, Toronto, ON, M4N 3M5, Canada
| | | |
Collapse
|
99
|
Jakobi A, Lühr A, Stützer K, Bandurska-Luque A, Löck S, Krause M, Baumann M, Perrin R, Richter C. Increase in Tumor Control and Normal Tissue Complication Probabilities in Advanced Head-and-Neck Cancer for Dose-Escalated Intensity-Modulated Photon and Proton Therapy. Front Oncol 2015; 5:256. [PMID: 26636038 PMCID: PMC4653282 DOI: 10.3389/fonc.2015.00256] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 11/06/2015] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Presently used radiochemotherapy regimens result in moderate local control rates for patients with advanced head-and-neck squamous cell carcinoma (HNSCC). Dose escalation (DE) may be an option to improve patient outcome, but may also increase the risk of toxicities in healthy tissue. The presented treatment planning study evaluated the feasibility of two DE levels for advanced HNSCC patients, planned with either intensity-modulated photon therapy (IMXT) or proton therapy (IMPT). MATERIALS AND METHODS For 45 HNSCC patients, IMXT and IMPT treatment plans were created including DE via a simultaneous integrated boost (SIB) in the high-risk volume, while maintaining standard fractionation with 2 Gy per fraction in the remaining target volume. Two DE levels for the SIB were compared: 2.3 and 2.6 Gy. Treatment plan evaluation included assessment of tumor control probabilities (TCP) and normal tissue complication probabilities (NTCP). RESULTS An increase of approximately 10% in TCP was estimated between the DE levels. A pronounced high-dose rim surrounding the SIB volume was identified in IMXT treatment. Compared to IMPT, this extra dose slightly increased the TCP values and to a larger extent the NTCP values. For both modalities, the higher DE level led only to a small increase in NTCP values (mean differences <2%) in all models, except for the risk of aspiration, which increased on average by 8 and 6% with IMXT and IMPT, respectively, but showed a considerable patient dependence. CONCLUSION Both DE levels appear applicable to patients with IMXT and IMPT since all calculated NTCP values, except for one, increased only little for the higher DE level. The estimated TCP increase is of relevant magnitude. The higher DE schedule needs to be investigated carefully in the setting of a prospective clinical trial, especially regarding toxicities caused by high local doses that lack a sound dose-response description, e.g., ulcers.
Collapse
Affiliation(s)
- Annika Jakobi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| | - Armin Lühr
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany ; German Cancer Consortium (DKTK), Partner Site Dresden , Dresden , Germany ; German Cancer Research Center (DKFZ) , Heidelberg , Germany
| | - Kristin Stützer
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| | - Anna Bandurska-Luque
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany ; Department of Radiation Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany ; German Cancer Consortium (DKTK), Partner Site Dresden , Dresden , Germany ; German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Radiation Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany ; Institute of Radiooncology, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| | - Michael Baumann
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany ; German Cancer Consortium (DKTK), Partner Site Dresden , Dresden , Germany ; German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Radiation Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany ; Institute of Radiooncology, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| | - Rosalind Perrin
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine, 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, University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany ; German Cancer Consortium (DKTK), Partner Site Dresden , Dresden , Germany ; German Cancer Research Center (DKFZ) , Heidelberg , Germany ; Department of Radiation Oncology, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden , Dresden , Germany ; Institute of Radiooncology, Helmholtz-Zentrum Dresden - Rossendorf , Dresden , Germany
| |
Collapse
|
100
|
Vera P, Thureau S. Nouvelles modalités d’imagerie pour la radiothérapie : imagerie fonctionnelle et moléculaire. Cancer Radiother 2015; 19:538-42. [DOI: 10.1016/j.canrad.2015.06.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
|