1
|
Yaromina A, Koi L, Schuitmaker L, van der Wiel AMMA, Dubois LJ, Krause M, Lambin P. Overcoming radioresistance with the hypoxia-activated prodrug CP-506: A pre-clinical study of local tumour control probability. Radiother Oncol 2023; 186:109738. [PMID: 37315579 DOI: 10.1016/j.radonc.2023.109738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND AND PURPOSE Tumour hypoxia is an established radioresistance factor. A novel hypoxia-activated prodrug CP-506 has been proven to selectively target hypoxic tumour cells and to cause anti-tumour activity. The current study investigates whether CP-506 improves outcome of radiotherapy in vivo. MATERIALS AND METHODS Mice bearing FaDu and UT-SCC-5 xenografts were randomized to receive 5 daily injections of CP-506/vehicle followed by single dose (SD) irradiation. In addition, CP-506 was combined once per week with fractionated irradiation (30 fractions/6 weeks). Animals were followed-up to score all recurrences. In parallel, tumours were harvested to evaluate pimonidazole hypoxia, DNA damage (γH2AX), expression of oxidoreductases. RESULTS CP-506 treatment significantly increased local control rate after SD in FaDu, 62% vs. 27% (p = 0.024). In UT-SCC-5, this effect was not curative and only marginally significant. CP-506 induced significant DNA damage in FaDu (p = 0.009) but not in UT- SCC-5. Hypoxic volume (HV) was significantly smaller (p = 0.038) after pretreatment with CP-506 as compared to vehicle in FaDu but not in less responsive UT-SCC-5. Adding CP-506 to fractionated radiotherapy in FaDu did not result in significant benefit. CONCLUSION The results support the use of CP-506 in combination with radiation in particular using hypofractionation schedules in hypoxic tumours. The magnitude of effect depends on the tumour model, therefore it is expected that applying appropriate patient stratification strategy will further enhance the benefit of CP-506 treatment for cancer patients. A phase I-IIA clinical trial of CP-506 in monotherapy or in combination with carboplatin or a checkpoint inhibitor has been approved (NCT04954599).
Collapse
Affiliation(s)
- Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands.
| | - Lydia Koi
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany
| | - Lesley Schuitmaker
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | | | - Ludwig Jerome Dubois
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiooncology-OncoRay, Dresden, Germany; German Cancer Consortium (DKTK), partner site Dresden, German Cancer Research Center, Heidelberg, National Center for Tumour Diseases (NCT), partner site Dresden, German Cancer Consortium (DKTK), core center Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philippe Lambin
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
2
|
Lan Y, Moustafa M, Knoll M, Xu C, Furkel J, Lazorchak A, Yeung TL, Hasheminasab SM, Jenkins MH, Meister S, Yu H, Schlegel J, Marelli B, Tang Z, Qin G, Klein C, Qi J, Zhou C, Locke G, Krunic D, Derner MG, Schwager C, Fontana RE, Kriegsmann K, Jiang F, Rein K, Kriegsmann M, Debus J, Lo KM, Abdollahi A. Simultaneous targeting of TGF-β/PD-L1 synergizes with radiotherapy by reprogramming the tumor microenvironment to overcome immune evasion. Cancer Cell 2021; 39:1388-1403.e10. [PMID: 34506739 DOI: 10.1016/j.ccell.2021.08.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 06/25/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022]
Abstract
Localized radiotherapy (RT) induces an immunogenic antitumor response that is in part counterbalanced by activation of immune evasive and tissue remodeling processes, e.g., via upregulation of programmed cell death-ligand 1 (PD-L1) and transforming growth factor β (TGF-β). We report that a bifunctional fusion protein that simultaneously inhibits TGF-β and PD-L1, bintrafusp alfa (BA), effectively synergizes with radiotherapy, leading to superior survival in multiple therapy-resistant murine tumor models with poor immune infiltration. The BA + RT (BART) combination increases tumor-infiltrating leukocytes, reprograms the tumor microenvironment, and attenuates RT-induced fibrosis, leading to reconstitution of tumor immunity and regression of spontaneous lung metastases. Consistently, the beneficial effects of BART are in part reversed by depletion of cytotoxic CD8+ T cells. Intriguingly, targeting of the TGF-β trap to PD-L1+ endothelium and the M2/lipofibroblast-like cell compartment by BA attenuated late-stage RT-induced lung fibrosis. Together, the results suggest that the BART combination has the potential to eradicate therapy-resistant tumors while sparing normal tissue, further supporting its clinical translation.
Collapse
Affiliation(s)
- Yan Lan
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany.
| | - Mahmoud Moustafa
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; Department of Clinical Pathology, Suez Canal University, Ismailia 41522, Egypt
| | - Maximilian Knoll
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Chunxiao Xu
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Jennifer Furkel
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Adam Lazorchak
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Tsz-Lun Yeung
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Sayed-Mohammad Hasheminasab
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; Department of Dermatology, Venereology and Allergology, Charité Universitätsmedizin, Berlin, Germany
| | - Molly H Jenkins
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Sarah Meister
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Huakui Yu
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Julian Schlegel
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Bo Marelli
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Zili Tang
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Guozhong Qin
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Carmen Klein
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Jin Qi
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Cheng Zhou
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - George Locke
- Department of Translational Medicine, EMD Serono Research & Development Institute, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Damir Krunic
- Light Microscopy Facility, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Melissa G Derner
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Christian Schwager
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Rachel E Fontana
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology, and Rheumatology, Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Feng Jiang
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Katrin Rein
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Mark Kriegsmann
- Department of Pathology, Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Juergen Debus
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany
| | - Kin-Ming Lo
- Department of TIP OIO, EMD Serono Research & Development Institute, 45 Middlesex Turnpike, Billerica, MA 01821, USA; an affiliate of Merck KGaA, Darmstadt, Germany
| | - Amir Abdollahi
- German Cancer Consortium (DKTK) Core-Center, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Divisions of Molecular & Translational Radiation Oncology, Heidelberg Faculty of Medicine (MFHD) and Heidelberg University Hospital (UKHD), Heidelberg Ion-Beam Therapy Center (HIT), 69120 Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany; CCU Translational Radiation Oncology, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ) and Heidelberg University Hospital (UKHD), 69120 Heidelberg, Germany.
| |
Collapse
|
3
|
Fan B, Li C, Mu F, Qin W, Wang L, Sun X, Wang C, Zou B, Wang S, Li W, Hu M. Dose escalation guided by 18F-FDG PET/CT for esophageal cancer. RADIATION MEDICINE AND PROTECTION 2021. [DOI: 10.1016/j.radmp.2021.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
4
|
Leger S, Zwanenburg A, Leger K, Lohaus F, Linge A, Schreiber A, Kalinauskaite G, Tinhofer I, Guberina N, Guberina M, Balermpas P, von der Grün J, Ganswindt U, Belka C, Peeken JC, Combs SE, Boeke S, Zips D, Richter C, Krause M, Baumann M, Troost EG, Löck S. Comprehensive Analysis of Tumour Sub-Volumes for Radiomic Risk Modelling in Locally Advanced HNSCC. Cancers (Basel) 2020; 12:cancers12103047. [PMID: 33086761 PMCID: PMC7589463 DOI: 10.3390/cancers12103047] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/13/2020] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Radiomic risk models are usually based on imaging features, which are extracted from the entire gross tumour volume (GTVentire). This approach does not explicitly consider the complex biological structure of the tumours. Therefore, in this retrospective study, we investigated the prognostic value of radiomic analyses based on different tumour sub-volumes using computed tomography imaging of patients with locally advanced head and neck squamous cell carcinoma who were treated with primary radio-chemotherapy. The GTVentire was cropped by different margins to define the rim and corresponding core sub-volumes of the tumour. Furthermore, the best performing tumour rim sub-volume was extended into surrounding tissue with different margins. As a result, the models based on the 5 mm tumour rim and on the 3 mm extended rim sub-volume showed an improved performance compared to models based on the corresponding tumour core. This indicates that the consideration of tumour sub-volumes may help to improve radiomic risk models. Abstract Imaging features for radiomic analyses are commonly calculated from the entire gross tumour volume (GTVentire). However, tumours are biologically complex and the consideration of different tumour regions in radiomic models may lead to an improved outcome prediction. Therefore, we investigated the prognostic value of radiomic analyses based on different tumour sub-volumes using computed tomography imaging of patients with locally advanced head and neck squamous cell carcinoma. The GTVentire was cropped by different margins to define the rim and the corresponding core sub-volumes of the tumour. Subsequently, the best performing tumour rim sub-volume was extended into surrounding tissue with different margins. Radiomic risk models were developed and validated using a retrospective cohort consisting of 291 patients in one of the six Partner Sites of the German Cancer Consortium Radiation Oncology Group treated between 2005 and 2013. The validation concordance index (C-index) averaged over all applied learning algorithms and feature selection methods using the GTVentire achieved a moderate prognostic performance for loco-regional tumour control (C-index: 0.61 ± 0.04 (mean ± std)). The models based on the 5 mm tumour rim and on the 3 mm extended rim sub-volume showed higher median performances (C-index: 0.65 ± 0.02 and 0.64 ± 0.05, respectively), while models based on the corresponding tumour core volumes performed less (C-index: 0.59 ± 0.01). The difference in C-index between the 5 mm tumour rim and the corresponding core volume showed a statistical trend (p = 0.10). After additional prospective validation, the consideration of tumour sub-volumes may be a promising way to improve prognostic radiomic risk models.
Collapse
Affiliation(s)
- Stefan Leger
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Correspondence:
| | - Alex Zwanenburg
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
| | - Karoline Leger
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Fabian Lohaus
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Annett Linge
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Andreas Schreiber
- Department of Radiotherapy, Hospital Dresden-Friedrichstadt, 01067 Dresden, Germany;
| | - Goda Kalinauskaite
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 10117 Berlin, Germany; (G.K.); (I.T.)
- Department of Radiooncology and Radiotherapy, Charité University Hospital, 10117 Berlin, Germany
| | - Inge Tinhofer
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 10117 Berlin, Germany; (G.K.); (I.T.)
- Department of Radiooncology and Radiotherapy, Charité University Hospital, 10117 Berlin, Germany
| | - Nika Guberina
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 45147 Essen, Germany; (N.G.); (M.G.)
- Department of Radiotherapy, University Hospital Essen, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Maja Guberina
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 45147 Essen, Germany; (N.G.); (M.G.)
- Department of Radiotherapy, University Hospital Essen, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany
| | - Panagiotis Balermpas
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 60596 Frankfurt, Germany; (P.B.); (J.v.d.G.)
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Jens von der Grün
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 60596 Frankfurt, Germany; (P.B.); (J.v.d.G.)
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Ute Ganswindt
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 81377 Munich, Germany; (U.G.); (C.B.); (J.C.P.); (S.E.C.)
- Department of Radiation Oncology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- Clinical Cooperation Group, Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, 81377 Munich, Germany
- Department of Radiation Oncology, Medical University of Innsbruck, Anichstraße 35, A-6020 Innsbruck, Austria
| | - Claus Belka
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 81377 Munich, Germany; (U.G.); (C.B.); (J.C.P.); (S.E.C.)
- Department of Radiation Oncology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
- Clinical Cooperation Group, Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, 81377 Munich, Germany
| | - Jan C. Peeken
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 81377 Munich, Germany; (U.G.); (C.B.); (J.C.P.); (S.E.C.)
- Department of Radiation Oncology, Technische Universität München, 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Stephanie E. Combs
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 81377 Munich, Germany; (U.G.); (C.B.); (J.C.P.); (S.E.C.)
- Department of Radiation Oncology, Technische Universität München, 81675 Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Simon Boeke
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 72076 Tübingen, Germany; (S.B.); (D.Z.)
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Daniel Zips
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 72076 Tübingen, Germany; (S.B.); (D.Z.)
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, 72076 Tübingen, Germany
| | - Christian Richter
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Mechthild Krause
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Michael Baumann
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Esther G.C. Troost
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Institute of Radiooncology—OncoRay, Helmholtz-Zentrum Dresden—Rossendorf, 01328 Dresden, Germany
| | - Steffen Löck
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden—Rossendorf, 01307 Dresden, Germany; (A.Z.); (K.L.); (F.L.); (A.L.); (C.R.); (M.K.); (M.B.); (E.G.C.T.); (S.L.)
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) Partner Site, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden of the German Cancer Research Center (DKFZ), Faculty of Medicine and University Hospital Carl Gustav Carus and Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
5
|
Duan C, Chaovalitwongse WA, Bai F, Hippe DS, Wang S, Thammasorn P, Pierce LA, Liu X, You J, Miyaoka RS, Vesselle HJ, Kinahan PE, Rengan R, Zeng J, Bowen SR. Sensitivity analysis of FDG PET tumor voxel cluster radiomics and dosimetry for predicting mid-chemoradiation regional response of locally advanced lung cancer. Phys Med Biol 2020; 65:205007. [PMID: 33027064 PMCID: PMC7593986 DOI: 10.1088/1361-6560/abb0c7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
We investigated the sensitivity of regional tumor response prediction to variability in voxel clustering techniques, imaging features, and machine learning algorithms in 25 patients with locally advanced non-small cell lung cancer (LA-NSCLC) enrolled on the FLARE-RT clinical trial. Metabolic tumor volumes (MTV) from pre-chemoradiation (PETpre) and mid-chemoradiation fluorodeoxyglucose-positron emission tomography (FDG PET) images (PETmid) were subdivided into K-means or hierarchical voxel clusters by standardized uptake values (SUV) and 3D-positions. MTV cluster separability was evaluated by CH index, and morphologic changes were captured by Dice similarity and centroid Euclidean distance. PETpre conventional features included SUVmean, MTV/MTV cluster size, and mean radiation dose. PETpre radiomics consisted of 41 intensity histogram and 3D texture features (PET Oncology Radiomics Test Suite) extracted from MTV or MTV clusters. Machine learning models (multiple linear regression, support vector regression, logistic regression, support vector machines) of conventional features or radiomic features were constructed to predict PETmid response. Leave-one-out-cross-validated root-mean-squared-error (RMSE) for continuous response regression (ΔSUVmean) and area-under-receiver-operating-characteristic-curve (AUC) for binary response classification were calculated. K-means MTV 2-clusters (MTVhi, MTVlo) achieved maximum CH index separability (Friedman p < 0.001). Between PETpre and PETmid, MTV cluster pairs overlapped (Dice 0.70-0.87) and migrated 0.6-1.1 cm. PETmid ΔSUVmean response prediction was superior in MTV and MTVlo (RMSE = 0.17-0.21) compared to MTVhi (RMSE = 0.42-0.52, Friedman p < 0.001). PETmid ΔSUVmean response class prediction performance trended higher in MTVlo (AUC = 0.83-0.88) compared to MTVhi (AUC = 0.44-0.58, Friedman p = 0.052). Models were more sensitive to MTV/MTV cluster regions (Friedman p = 0.026) than feature sets/algorithms (Wilcoxon signed-rank p = 0.36). Top-ranked radiomic features included GLZSM-LZHGE (large-zone-high-SUV), GTSDM-CP (cluster-prominence), GTSDM-CS (cluster-shade) and NGTDM-CNT (contrast). Top-ranked features were consistent between MTVhi and MTVlo cluster pairs but varied between MTVhi-MTVlo clusters, reflecting distinct regional radiomic phenotypes. Variability in tumor voxel cluster response prediction can inform robust radiomic target definition for risk-adaptive chemoradiation in patients with LA-NSCLC. FLARE-RT trial: NCT02773238.
Collapse
Affiliation(s)
- Chunyan Duan
- Department of Mechanical Engineering, Tongji University School of Mechanical Engineering, Shanghai China
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - W. Art Chaovalitwongse
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Fangyun Bai
- Department of Management Science and Engineering, Tongji University School of Economics and Management, Shanghai China
- Department of Industrial, Manufacturing, & Systems Engineering, University of Texas at Arlington College of Engineering, Arlington, TX
| | - Daniel S. Hippe
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Shouyi Wang
- Department of Industrial, Manufacturing, & Systems Engineering, University of Texas at Arlington College of Engineering, Arlington, TX
| | - Phawis Thammasorn
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Larry A. Pierce
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Xiao Liu
- Department of Industrial Engineering, University of Arkansas College of Engineering, Fayetteville AR
| | - Jianxin You
- Department of Management Science and Engineering, Tongji University School of Economics and Management, Shanghai China
| | - Robert S. Miyaoka
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Hubert J. Vesselle
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Paul E. Kinahan
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| | - Ramesh Rengan
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - Jing Zeng
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
| | - Stephen R. Bowen
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle WA
- Department of Radiology, University of Washington School of Medicine, Seattle WA
| |
Collapse
|
6
|
Roberts PR, Jani AB, Packianathan S, Albert A, Bhandari R, Vijayakumar S. Upcoming imaging concepts and their impact on treatment planning and treatment response in radiation oncology. Radiat Oncol 2018; 13:146. [PMID: 30103786 PMCID: PMC6088418 DOI: 10.1186/s13014-018-1091-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
For 2018, the American Cancer Society estimated that there would be approximately 1.7 million new diagnoses of cancer and about 609,640 cancer-related deaths in the United States. By 2030 these numbers are anticipated to exceed a staggering 21 million annual diagnoses and 13 million cancer-related deaths. The three primary therapeutic modalities for cancer treatments are surgery, chemotherapy, and radiation therapy. Individually or in combination, these treatment modalities have provided and continue to provide curative and palliative care to the myriad victims of cancer. Today, CT-based treatment planning is the primary means through which conventional photon radiation therapy is planned. Although CT remains the primary treatment planning modality, the field of radiation oncology is moving beyond the sole use of CT scans to define treatment targets and organs at risk. Complementary tissue scans, such as magnetic resonance imaging (MRI) and positron electron emission (PET) scans, have all improved a physician’s ability to more specifically identify target tissues, and in some cases, international guidelines have even been issued. Moreover, efforts to combine PET and MR to define solid tumors for radiotherapy planning and treatment evaluation are also gaining traction. Keeping these advances in mind, we present brief overviews of other up-and-coming key imaging concepts that appear promising for initial treatment target definition or treatment response from radiation therapy.
Collapse
Affiliation(s)
- Paul Russell Roberts
- Department of Radiation Oncology, University of Mississippi Medical Center, 350 Woodrow Wilson Drive Suite 1600, Jackson, MS, 39213, USA
| | - Ashesh B Jani
- Department of Radiation Oncology, Winship Cancer Institute of Emory University, 1365 Clifton Rd, Atlanta, GA, 30322, USA
| | - Satyaseelan Packianathan
- Department of Radiation Oncology, University of Mississippi Medical Center, 350 Woodrow Wilson Drive Suite 1600, Jackson, MS, 39213, USA
| | - Ashley Albert
- Department of Radiation Oncology, University of Mississippi Medical Center, 350 Woodrow Wilson Drive Suite 1600, Jackson, MS, 39213, USA
| | - Rahul Bhandari
- Department of Radiation Oncology, University of Mississippi Medical Center, 350 Woodrow Wilson Drive Suite 1600, Jackson, MS, 39213, USA
| | - Srinivasan Vijayakumar
- Department of Radiation Oncology, University of Mississippi Medical Center, 350 Woodrow Wilson Drive Suite 1600, Jackson, MS, 39213, USA.
| |
Collapse
|
7
|
Fan B, Fan P, Kong L, Sun X, Zhao S, Sun X, Fu Z, Zheng J, Ma L, Wang S, Hu M, Yu J. 18F-deoxyglucose positron emission tomography/computed tomography to predict local failure in esophageal squamous cell carcinoma. Oncotarget 2018; 8:34498-34506. [PMID: 28404900 PMCID: PMC5470985 DOI: 10.18632/oncotarget.15606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/04/2016] [Indexed: 01/24/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) patients are at risk for local failure (LF) following treatment. Predicting tumor regions at high risk for local failure before radiotherapy may increase treatment efficacy by permitting an escalated radiation dose specifically to those regions critical for tumor control. Forty-one patients with non-resectable locally advanced ESCC underwent 18F-deoxyglucose positron emission tomography/computed tomography (FDG PET/CT) imaging before concurrent chemoradiotherapy (CCRT). After CCRT, a second (failure) FDG PET/CT was performed in cases of relapse. Failure FDG PET/CT scans were fused to pre-treatment scans to identify tumor regions at high risk for LF. Within a median follow-up time of 26 months, 20 patients (48.8%) had LF. In 19 patients, the failure occurred within a pre-treatment high FDG uptake region; the failure occurred outside these regions in only one patient. Pre-treatment metabolic tumor volume (MTV) was independently associated with LF (P<0.001, HR 1.128, 95% CI: 1.061–1.198). LF was more likely in patients with MTVs ≥27 cm3. In initial PET/CT images, when 50% maximum standardized uptake value (SUVmax) was used as the threshold, delineated subvolumes overlapped LF regions. These results confirm that LF occurs most commonly within pre-treatment high FDG uptake regions.
Collapse
Affiliation(s)
- Bingjie Fan
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Pingping Fan
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Li Kong
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Xindong Sun
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Shuqiang Zhao
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Nuclear Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Xiaorong Sun
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Nuclear Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Zheng Fu
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Nuclear Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Jinsong Zheng
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Nuclear Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Li Ma
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Nuclear Medicine, Shandong Cancer Hospital and Institute, Jinan, China
| | - Shijiang Wang
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Man Hu
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| | - Jinming Yu
- Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China.,Shandong Academy of Medical Sciences, Jinan, China.,Departments of Radiation Oncology and Shandong Province Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, China
| |
Collapse
|
8
|
Dębiec K, Wydmański J, Gorczewska I, Leszczyńska P, Gorczewski K, Leszczyński W, d’Amico A, Kalemba M. 18-Fluorodeoxy-Glucose Positron Emission Tomography- Computed Tomography (18-FDG-PET/CT) for Gross Tumor Volume (GTV) Delineation in Gastric Cancer Radiotherapy. Asian Pac J Cancer Prev 2017; 18:2989-2998. [PMID: 29172270 PMCID: PMC5773782 DOI: 10.22034/apjcp.2017.18.11.2989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose: Evaluation of the 18-fluorodeoxy-glucose positron emission tomography-computed tomography (18-FDG-PET/CT) for gross tumor volume (GTV) delineation in gastric cancer patients undergoing radiotherapy. Methods: In this study, 29 gastric cancer patients (17 unresectable and 7 inoperable) were initially enrolled for radical chemoradiotherapy (45Gy/25 fractions + chemotherapy based on 5 fluorouracil) or radiotherapy alone (45Gy/25 fractions) with planning based on the 18-FDG-PET/CT images. Five patients were excluded due to excess blood glucose levels (1), false-negative positron emission tomography (1) and distant metastases revealed by 18-FDG-PET/CT (3). The analysis involved measurement of metabolic tumor volumes (MTVs) performed on PET/CT workstations. Different threshold levels of the standardized uptake value (SUV) and liver uptake were set to obtain MTVs. Secondly, GTVPET values were derived manually using the positron emission tomography (PET) dataset blinded to the computed tomography (CT) data. Subsequently, GTVCT values were delineated using a radiotherapy planning system based on the CT scans blinded to the PET data. The referenced GTVCT values were correlated with the GTVPET and were compared with a conformality index (CI). Results: The mean CI was 0.52 (range, 0.12-0.85). In 13/24 patients (54%), the GTVPET was larger than GTVCT, and in the remainder, GTVPET was smaller. Moreover, the cranio-caudal diameter of GTVPET in 16 cases (64%) was larger than that of GTVCT, smaller in 7 cases (29%), and unchanged in one case. Manual PET delineation (GTVPET) achieved the best correlation with GTVCT (Pearson correlation = 0.76, p <0.0001). Among the analyzed MTVs, a statistically significant correlation with GTVCT was revealed for MTV10%SUVmax (r = 0.63; p = 0.0014), MTVliv (r = 0.60; p = 0.0021), MTVSUV2.5 (r = 0.54; p = 0.0063); MTV20%SUVmax (r = 0.44; p = 0.0344); MTV30%SUVmax (r = 0.44; p = 0.0373). Conclusion: 18-FDG-PET/CT in gastric cancer radiotherapy planning may affect the GTV delineation.
Collapse
Affiliation(s)
- Kinga Dębiec
- Radiotherapy and Chemotherapy I Clinic, Maria Skłodowska-Curie Memorial Institute of Oncology, Gliwice Branch. Poland.
| | | | | | | | | | | | | | | |
Collapse
|
9
|
A novel concept for tumour targeting with radiation: Inverse dose-painting or targeting the “Low Drug Uptake Volume”. Radiother Oncol 2017; 124:513-520. [DOI: 10.1016/j.radonc.2017.04.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/17/2017] [Accepted: 04/21/2017] [Indexed: 01/21/2023]
|
10
|
Rostami A, Bratman SV. Utilizing circulating tumour DNA in radiation oncology. Radiother Oncol 2017; 124:357-364. [DOI: 10.1016/j.radonc.2017.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 07/01/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022]
|
11
|
Jentsch C, Bergmann R, Brüchner K, Mosch B, Yaromina A, Krause M, Zips D, Troost EG, Löck S, Kotzerke J, Steinbach J, Thames H, Baumann M, Beuthien-Baumann B. Impact of pre- and early per-treatment FDG-PET based dose-escalation on local tumour control in fractionated irradiated FaDu xenograft tumours. Radiother Oncol 2016; 121:447-452. [DOI: 10.1016/j.radonc.2016.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/16/2016] [Accepted: 07/28/2016] [Indexed: 10/20/2022]
|
12
|
Baumann M, Krause M, Overgaard J, Debus J, Bentzen SM, Daartz J, Richter C, Zips D, Bortfeld T. Radiation oncology in the era of precision medicine. Nat Rev Cancer 2016; 16:234-49. [PMID: 27009394 DOI: 10.1038/nrc.2016.18] [Citation(s) in RCA: 562] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Technological advances and clinical research over the past few decades have given radiation oncologists the capability to personalize treatments for accurate delivery of radiation dose based on clinical parameters and anatomical information. Eradication of gross and microscopic tumours with preservation of health-related quality of life can be achieved in many patients. Two major strategies, acting synergistically, will enable further widening of the therapeutic window of radiation oncology in the era of precision medicine: technology-driven improvement of treatment conformity, including advanced image guidance and particle therapy, and novel biological concepts for personalized treatment, including biomarker-guided prescription, combined treatment modalities and adaptation of treatment during its course.
Collapse
Affiliation(s)
- Michael Baumann
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Oncology, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Mechthild Krause
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiation Oncology, Bautzner Landstrasse 400, 01328 Dresden, Germany
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Nørrebrogade 44, 8000 Aarhus C, Denmark
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Im Neuenheimer Feld 460, 69120 Heidelberg
- Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Im Neuenheimer Feld 400, 69120 Heidelberg
- German Cancer Consortium (DKTK) Heidelberg, Germany
| | - Søren M Bentzen
- Department of Epidemiology and Public Health and Greenebaum Cancer Center, University of Maryland School of Medicine, 22 S Greene Street S9a03, Baltimore, Maryland 21201, USA
| | - Juliane Daartz
- Department of Radiation Oncology, Physics Division, Massachusetts General Hospital and Harvard Medical School, 1000 Blossom Street Cox 362, Boston, Massachusetts 02114, USA
| | - Christian Richter
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden
- OncoRay - National Center for Radiation Research in Oncology (NCRO), Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, and Helmholtz-Zentrum Dresden-Rossendorf, Fetscherstrasse 74, 01307 Dresden
- National Center for Tumor Diseases (NCT), Fetscherstrasse 74, 01307 Dresden
- German Cancer Consortium (DKTK) Dresden, Germany
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Daniel Zips
- German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
- German Cancer Consortium Tübingen, Postfach 2669, 72016 Tübingen
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, Hoppe-Seyler-Strasse 3, 72016 Tübingen, Germany
| | - Thomas Bortfeld
- Department of Radiation Oncology, Physics Division, Massachusetts General Hospital and Harvard Medical School, 1000 Blossom Street Cox 362, Boston, Massachusetts 02114, USA
| |
Collapse
|
13
|
Knudtsen IS, Svestad JG, Skaug Sande EP, Rekstad BL, Rødal J, van Elmpt W, Öllers M, Hole EO, Malinen E. Validation of dose painting of lung tumours using alanine/EPR dosimetry. Phys Med Biol 2016; 61:2243-54. [DOI: 10.1088/0031-9155/61/6/2243] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
14
|
Fliedner FP, Hansen AE, Jørgensen JT, Kjær A. The use of matrigel has no influence on tumor development or PET imaging in FaDu human head and neck cancer xenografts. BMC Med Imaging 2016; 16:5. [PMID: 26762341 PMCID: PMC4712613 DOI: 10.1186/s12880-016-0105-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/04/2016] [Indexed: 11/10/2022] Open
Abstract
Background In preclinical research MatrixgelTM Basement Membrane Matrix (MG) is used frequently for the establishment of syngeneic and xenograft cancer models. Limited information on its influence on parameters including; tumor growth, vascularization, hypoxia and imaging characteristics is currently available. This study evaluates the potential effect of matrigel use in a human head and neck cancer xenograft model (FaDu; hypopharyngeal carcinoma) in NMRI nude mice. The FaDu cell line was chosen based on its frequent use in studies of cancer imaging and tumor microenvironment. Methods NMRI nude mice (n = 34) were divided into two groups and subcutaneously injected with FaDu cells in medium either including (+MG) or excluding matrigel (−MG). In sub study I seven mice from each group (+MG, n = 7; −MG, n = 7) were 18F- fluorodeoxyglucose (18F-FDG) PET/CT scanned on Day 5, 8, 12, 15, and 19. In sub study II ten mice from each group (+MG, n = 10; −MG, n = 10) were included and tumors collected for immunohistochemistry (IHC) analysis of tumor microenvironment including; proliferation ratio, micro vessel density, average vessel area, hypoxia, nuclear density, and necrosis. Tumors for IHC were collected according to size (200–400 mm3, 500–700 mm3, 800–1100 mm3). Results FDG uptake and tumor growth was statistically compatible for the tumors established with or without MG. The IHC analysis on all parameters only identified a significantly higher micro vessel density for tumor size 500–700 mm3 and 800–1100 mm3 and average vessel area for tumor size 500–700 mm3 in the −MG group. Comparable variations were observed for tumors of both the +MG and −MG groups. No difference in tumor take rate was observed between groups in study. Conclusions Matrigel did not affect tumor growth or tumor take for the FaDu xenograft model evaluated. Tumors in the -MG group displayed increased angiogenesis compared to the +MG tumors. No difference in 18F-FDG PET uptake for tumors of different groups was found. Based on these observations the influence of matrigel on tumor imaging and tumor microenvironment seems minor for this particular xenograft model.
Collapse
Affiliation(s)
- Frederikke P Fliedner
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Anders E Hansen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark. .,Department of Micro- and Nanotechnology, Center for Nanomedicine and Theranostics, DTU Nanotech, Technical University of Denmark, Building 423, 2800, Lyngby, Denmark.
| | - Jesper T Jørgensen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| |
Collapse
|
15
|
Preclinical Assessment of Efficacy of Radiation Dose Painting Based on Intratumoral FDG-PET Uptake. Clin Cancer Res 2015; 21:5511-8. [DOI: 10.1158/1078-0432.ccr-15-0290] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/03/2015] [Indexed: 11/16/2022]
|
16
|
Jentsch C, Beuthien-Baumann B, Troost EGC, Shakirin G. Validation of functional imaging as a biomarker for radiation treatment response. Br J Radiol 2015; 88:20150014. [PMID: 26083533 DOI: 10.1259/bjr.20150014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Major advances in radiotherapy techniques, increasing knowledge of tumour biology and the ability to translate these advances into new therapeutic approaches are important goals towards more individualized cancer treatment. With the development of non-invasive functional and molecular imaging techniques such as positron emission tomography (PET)-CT scanning and MRI, there is now a need to evaluate potential new biomarkers for tumour response prediction, for treatment individualization is not only based on morphological criteria but also on biological tumour characteristics. The goal of individualization of radiotherapy is to improve treatment outcome and potentially reduce chronic treatment toxicity. This review gives an overview of the molecular and functional imaging modalities of tumour hypoxia and tumour cell metabolism, proliferation and perfusion as predictive biomarkers for radiation treatment response in head and neck tumours and in lung tumours. The current status of knowledge on integration of PET/CT/MRI into treatment management and bioimage-guided adaptive radiotherapy are discussed.
Collapse
Affiliation(s)
- C Jentsch
- 1 OncoRay-National Centre for Radiation Research in Oncology, Dresden, Germany.,2 Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden,Helmholtz-Zentrum Dresden-Rossendorf, Germany.,3 German Cancer Consortium (DKTK) Dresden, Germany
| | - B Beuthien-Baumann
- 1 OncoRay-National Centre for Radiation Research in Oncology, Dresden, Germany.,3 German Cancer Consortium (DKTK) Dresden, Germany.,4 Institute of Radiation Oncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - E G C Troost
- 1 OncoRay-National Centre for Radiation Research in Oncology, Dresden, Germany.,2 Department of Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden,Helmholtz-Zentrum Dresden-Rossendorf, Germany.,3 German Cancer Consortium (DKTK) Dresden, Germany.,4 Institute of Radiation Oncology, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | | |
Collapse
|
17
|
van Dijk LK, Boerman OC, Franssen GM, Lok J, Kaanders JHAM, Bussink J. Early response monitoring with 18F-FDG PET and cetuximab-F(ab')2-SPECT after radiotherapy of human head and neck squamous cell carcinomas in a mouse model. J Nucl Med 2014; 55:1665-70. [PMID: 25236350 DOI: 10.2967/jnumed.114.141762] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Only a subset of patients with head and neck squamous cell carcinomas (HNSCCs) benefit from radiotherapy and concurrent epidermal growth factor receptor (EGFR) inhibitor therapy with cetuximab, indicating the need for patient selection. The aim of this study was to visualize the change in systemically accessible EGFR with (111)In-cetuximab-F(ab')2 SPECT before and after radiotherapy, while simultaneously evaluating (18)F-FDG PET uptake. METHODS Mice with HNSCC xenografts, cetuximab-sensitive SCCNij202 and cetuximab-resistant SCCNij167, were imaged with SPECT/CT using (111)In-cetuximab-F(ab')2 as a tracer, directly followed by PET imaging with (18)F-FDG. Scans were acquired 7 d before radiotherapy (10 Gy) and 1, 7, and 14 d after treatment. Intratumoral localization of (111)In-cetuximab-F(ab')(2) was evaluated by autoradiography and histologic markers evaluated by immunofluorescence staining in the same tumor sections. RESULTS Growth of irradiated SCCNij202 and SCCNij167 tumors was significantly delayed, compared with controls (P < 0.05). No changes in uptake of (18)F-FDG were observed in either of the xenografts after radiotherapy. SPECT images of tumor-bearing mice showed a significant increase in uptake of (111)In-cetuximab-F(ab')(2) in the SCCNij202 tumors after irradiation (tumor-to-liver ratio, 4.3 ± 1.1 vs. 10.5 ± 3.3, 7 d before and 14 d after treatment, respectively, P < 0.01) but not in SCCNij167 tumors. Immunohistochemical EGFR staining showed a translocation of the EGFR from the cytoplasm to the cell membrane in irradiated SCCNij202 xenografts. Intratumoral distribution of (111)In-cetuximab-F(ab')(2) as determined by autoradiography correlated well with the distribution of EGFR as determined immunohistochemically (r = 0.85; range, 0.69-0.95). CONCLUSION EGFR accessibility can be visualized with (111)In-cetuximab-F(ab')(2). (111)In-cetuximab-F(ab')(2) uptake increased after irradiation only in cetuximab-sensitive SCCNij202 xenografts, implying that the tracer can be used to measure irradiation-induced changes of EGFR expression and can monitor the compensatory response of tumors to radiotherapy.
Collapse
Affiliation(s)
- Laura K van Dijk
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; and Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jasper Lok
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Johannes H A M Kaanders
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; and
| | - Johan Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; and
| |
Collapse
|
18
|
Wilson JM, Partridge M, Hawkins M. The application of functional imaging techniques to personalise chemoradiotherapy in upper gastrointestinal malignancies. Clin Oncol (R Coll Radiol) 2014; 26:581-96. [PMID: 24998430 PMCID: PMC4150923 DOI: 10.1016/j.clon.2014.06.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Revised: 05/22/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
Abstract
Functional imaging gives information about physiological heterogeneity in tumours. The utility of functional imaging tests in providing predictive and prognostic information after chemoradiotherapy for both oesophageal cancer and pancreatic cancer will be reviewed. The benefit of incorporating functional imaging into radiotherapy planning is also evaluated. In cancers of the upper gastrointestinal tract, the vast majority of functional imaging studies have used (18)F-fluorodeoxyglucose positron emission tomography (FDG-PET). Few studies in locally advanced pancreatic cancer have investigated the utility of functional imaging in risk-stratifying patients or aiding target volume definition. Certain themes from the oesophageal data emerge, including the need for a multiparametric assessment of functional images and the added value of response assessment rather than relying on single time point measures. The sensitivity and specificity of FDG-PET to predict treatment response and survival are not currently high enough to inform treatment decisions. This suggests that a multimodal, multiparametric approach may be required. FDG-PET improves target volume definition in oesophageal cancer by improving the accuracy of tumour length definition and by improving the nodal staging of patients. The ideal functional imaging test would accurately identify patients who are unlikely to achieve a pathological complete response after chemoradiotherapy and would aid the delineation of a biological target volume that could be used for treatment intensification. The current limitations of published studies prevent integrating imaging-derived parameters into decision making on an individual patient basis. These limitations should inform future trial design in oesophageal and pancreatic cancers.
Collapse
Affiliation(s)
- J M Wilson
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, UK.
| | - M Partridge
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - M Hawkins
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Oxford, UK
| |
Collapse
|
19
|
Son SH, Kim DH, Hong CM, Kim CY, Jeong SY, Lee SW, Lee J, Ahn BC. Prognostic implication of intratumoral metabolic heterogeneity in invasive ductal carcinoma of the breast. BMC Cancer 2014; 14:585. [PMID: 25112709 PMCID: PMC4148559 DOI: 10.1186/1471-2407-14-585] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 08/05/2014] [Indexed: 11/16/2022] Open
Abstract
Background The purpose of this study was to evaluate the prognostic implication of findings of intratumoral metabolic heterogeneity on pretreatment 18F-FDG PET/CT scans in patients with invasive ductal carcinoma (IDC) of the breast. Methods One hundred and twenty-three female IDC patients who underwent pretreatment 18F-fluorodeoxyglucose positron-emission tomography/computed tomography (18F-FDG PET/CT) scans were retrospectively evaluated in this study. The heterogeneity factor (HF) defined as the derivative (dV/dT) of a volume threshold function from 40% to 80%, was computed for each primary tumor. Other metabolic PET parameters (maximum standardized uptake value [SUVmax], metabolic tumor volume [MTV], and total lesion glycolysis [TLG]) were measured. The HF was compared with clinicopathologic factors and other PET parameters. Univariate and multivariate analyses for the overall survival (OS) were performed. Results The HF ranged from 0.02 to 6.72 (mean, 0.35 ± 0.82) and significantly correlated with MTV (r = 0.955; p < 0.0001) and TLG (r = 0.354; p = 0.0001). The HF was significantly higher (implying more heterogeneity) in tumors with higher T and N stages. The optimal cut-off values for the OS determined using a receiver operating characteristic (ROC) curve were 0.34 for the HF, 5.6 for SUVmax, 8.55 cm3 for MTV, and 14.43 for TLG. The OS rate among the 123 patients was 86.2%. T stage (1, 2 vs. 3, 4), N stage (0, 1 vs. 2, 3), M stage (0 vs. 1), ER status (+ vs. –), SUVmax (≤ 5.6 vs. > 5.6), MTV (≤ 8.55 cm3 vs. > 8.55 cm3), TLG (≤ 14.43 vs. > 14.43), and HF (< 0.34 vs. ≥ 0.34) affected the OS on univariate analysis. After adjustment for the effects of TNM stage and ER status, the HF and MTV were significant predictors of OS. Among the PET parameters, the best prognostic factor for OS was the HF. Conclusions Intratumoral metabolic heterogeneity correlated closely with the MTV and significantly affected the OS in IDC patients. The HF may act as a robust surrogate marker for the prediction of OS in IDC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, Kyungpook National University School of Medicine and Hospital, 50 Samduk-dong 2-ga, Jung-gu, Daegu 700-721, Republic of Korea.
| |
Collapse
|
20
|
Wilson JM, Mukherjee S, Chu KY, Brunner TB, Partridge M, Hawkins M. Challenges in using ¹⁸F-fluorodeoxyglucose-PET-CT to define a biological radiotherapy boost volume in locally advanced pancreatic cancer. Radiat Oncol 2014; 9:146. [PMID: 24962658 PMCID: PMC4078370 DOI: 10.1186/1748-717x-9-146] [Citation(s) in RCA: 13] [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: 03/07/2014] [Accepted: 06/10/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The best method of identifying regions within pancreatic tumours that might benefit from an increased radiotherapy dose is not known. We investigated the utility of pre-treatment FDG-PET in predicting the spatial distribution of residual metabolic activity following chemoradiotherapy (CRT) in locally advanced pancreatic cancer (LAPC). METHODS 17 patients had FDG-PET/CT scans at baseline and six weeks post-CRT. Tumour segmentation was performed at 40% and 50% of SUVmax at baseline and 60%, 70%, 80% and 90% post-CRT. FDG-PET scans were non-rigidly registered to the radiotherapy planning CT using the CT component of the FDG-PET/CT. Percentage overlap of the post-CRT volumes with the pre-CRT volumes with one another and the gross tumour volume (GTV) was calculated. RESULTS SUVmax decreased during CRT (median pre- 8.0 and post- 3.6, p < 0.0001). For spatial correlation analysis, 9 pairs of scans were included (Four were excluded following complete metabolic response, one patient had a non-FDG avid tumour, one had no post-CRT imaging, one had diffuse FDG uptake that could not be separated from normal tissues and one had an elevated blood glucose). The Pre40% and 50% of SUVmax volumes covered a mean of 50.8% and 30.3% of the GTV respectively. The mean% overlap of the 90%, 80%, 70%, 60% of SUVmax post-CRT with the Pre40% and Pre50% volumes were 83.3%, 84.0%, 83.7%, 77.9% and 77.8%, 69.9%, 74.5%, 64.8% respectively. CONCLUSIONS Regions of residual metabolic activity following CRT can be predicted from the baseline FDG-PET and could aid definition of a biological target volume for non-uniform dose prescriptions.
Collapse
Affiliation(s)
- James M Wilson
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Somnath Mukherjee
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Kwun-Ye Chu
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Thomas B Brunner
- Department of Radiation Oncology, University Hospitals of Freiburg, Robert-Koch-Str. 3, D-79106 Freiburg im Breisgau, Germany
| | - Mike Partridge
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX3 7DQ, UK
| | - Maria Hawkins
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford OX3 7DQ, UK
| |
Collapse
|
21
|
Jeong J, Setton JS, Lee NY, Oh JH, Deasy JO. Estimate of the impact of FDG-avidity on the dose required for head and neck radiotherapy local control. Radiother Oncol 2014; 111:340-7. [PMID: 24833560 PMCID: PMC4822492 DOI: 10.1016/j.radonc.2014.03.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 03/14/2014] [Accepted: 03/22/2014] [Indexed: 11/29/2022]
Abstract
Background and purpose Although FDG-avid tumors are recognized as a potential target for dose escalation, there is no clear basis for selecting a boost dose to counter this apparent radioresistance. Using a novel analysis method, based on the new concept of an outcome-equivalent dose, we estimate the extra dose required to equalize local control between FDG-avid and non-avid head and neck tumors. Materials and methods Based on a literature review, five reports of head and neck cancer (423 patients in total), along with an internal validation dataset from our institution (135 oropharynx patients), were used in this analysis. To compensate for the heterogeneity among multi-institutional patient cohorts and corresponding treatment techniques, local control data of the cohorts were fit to a single dose–response curve with a clinically representative steepness (γ50 = 2), thereby defining an ‘outcome-equivalent dose’ (OED) for each institutional cohort. Separate dose–response curves were then determined for the FDG-avid and FDG-non-avid patient cohorts, and the ratio of TD50 (tumor dose required for 50% of control) values between the high- and low-FDG-uptake groups (TD50,high/TD50,low) was estimated, resulting in an estimated metabolic dose-modifying factor (mDMF) due to FDG-avidity. Results For individual datasets, the estimated mDMFs were found to be in the range of 1.07–1.62, decreasing if the assumed slope (γ50) increased. Weighted logistic regression for the six datasets resulted in a mDMF of 1.19 [95% CI: 1.04–1.34] for a γ50 value of 2, which translates to a needed dose increase of about 1.5 Gy per unit increase in the maximum standardized uptake value (SUVm) of FDG-PET [95% CI: 0.3–2.7]. Assumptions of lower or higher γ50 values (1.5 or 2.5) resulted in slightly different mDMFs: 1.26 or 1.15, respectively. A validation analysis with seven additional datasets, based on relaxed criteria, was consistent with the estimated mDMF. Conclusions We introduced a novel outcome-equivalent dose analysis method to estimate the dose– response modifying effect of FDG uptake variation. To reach equal response rates, FDG-avid tumors are likely to require 10% to 30% more dose than FDG-non-avid tumors. These estimates provide a rational starting point for selecting IMRT boosts for FDG-avid tumors. However, independent tests and refinements of the estimated dose-modifying effect, using high-quality prospective clinical trial data, are needed.
Collapse
Affiliation(s)
- Jeho Jeong
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Jeremy S Setton
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, USA.
| |
Collapse
|
22
|
Helbig L, Yaromina A, Sriramareddy SN, Böke S, Koi L, Thames HD, Baumann M, Zips D. Prognostic value of HIF-1α expression during fractionated irradiation. Strahlenther Onkol 2012; 188:1031-7. [PMID: 23053140 DOI: 10.1007/s00066-012-0150-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/18/2012] [Indexed: 01/12/2023]
Abstract
BACKGROUND AND PURPOSE Hypoxia and reoxygenation are important determinants of outcome after radiotherapy. HIF-1α is a key molecule involved in cellular response to hypoxia. HIF-1α expression levels have been shown to change after irradiation. The objective of the present study was to explore the prognostic value of HIF-1α expression during fractionated irradiation. MATERIALS AND METHODS Six human squamous cell carcinoma models xenografted in nude mice were analysed. Tumours were excised after 3, 5 and 10 fractions. HIF-1α expression was quantified by western blot. For comparative analysis, previously published data on local tumour control data and pimonidazole hypoxic fraction was used. RESULTS HIF-1α expression in untreated tumours exhibited intertumoural heterogeneity and did not correlate with pimonidazole hypoxic fraction. During fractionated irradiation the majority of tumour models exhibited a decrease in HIF-1α expression, whereas in UT-SCC-5 no change was observed. Neither kinetics nor expression levels during fractionated irradiation correlated with local tumour control. CONCLUSION Our data do not support the use of HIF-1α determined during treatment as a biomarker to predict outcome after fractionated irradiation.
Collapse
Affiliation(s)
- L Helbig
- Dept. of Radiation Oncology/ OncoRay National Center for Radiation Research, Medical Faculty and University Hospital Carl Gustav Carus Technische Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Huang B, Chan T, Kwong DLW, Chan WKS, Khong PL. Nasopharyngeal carcinoma: investigation of intratumoral heterogeneity with FDG PET/CT. AJR Am J Roentgenol 2012; 199:169-174. [PMID: 22733909 DOI: 10.2214/ajr.11.7336] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
OBJECTIVE The purpose of this study was to quantitatively evaluate the role of intratumoral heterogeneity of (18)F-FDG uptake in characterizing nasopharyngeal carcinoma (NPC). SUBJECTS AND METHODS Forty consecutively registered patients with newly diagnosed NPC underwent PET/CT. The heterogeneity factor, defined as the derivative of a volume threshold function, was computed for each tumor. The relations between heterogeneity factor and maximum standardized uptake value (SUV(max)), tumor volume, and TNM category were determined by two-tailed Spearman correlation. Factors that potentially affect outcome determined by disease-free survival were studied by Kaplan-Meier analysis with a log-rank test for univariate analysis and the Cox proportional hazard model for multivariate analysis. RESULTS The heterogeneity factor ranged from -1.80 to -0.13 (mean, -0.40 [SD, 0.40]) and significantly correlated with SUV(max) (r = -0.372; p = 0.018), tumor volume (r = -0.983; p < 0.001), and T category (r = -0.457; p = 0.003) but not with N and M categories. There was a significant difference in heterogeneity factor between T1 and T2 tumors and T3 and T4 tumors (p = 0.012). The 2-year disease-free survival rate among the 38 patients was 67.4%. According to the results of Kaplan-Meier analysis with the log-rank test, heterogeneity factor and M category significantly affected disease-free survival. Patients with tumors that had a heterogeneity factor greater than -0.24 (less-heterogeneous group) (p = 0.0498) or M0 status (p < 0.001) had better disease-free survival rates. Multivariate analysis showed only M category to be an independent predictor of disease-free survival (p < 0.001). CONCLUSION The intratumoral heterogeneity of FDG uptake varies across NPC tumors, significantly correlates with tumor aggressiveness, and is predictive of patient outcome. These findings may be useful for characterizing NPC, predicting survival, and improving patient care.
Collapse
Affiliation(s)
- Bingsheng Huang
- Department of Diagnostic Radiology, University of Hong Kong, Queen Mary Hospital, Rm 406, Block K, 102 Pokfulam Rd, Hong Kong, China
| | | | | | | | | |
Collapse
|
24
|
Kniess T, Laube M, Bergmann R, Sehn F, Graf F, Steinbach J, Wuest F, Pietzsch J. Radiosynthesis of a ¹⁸F-labeled 2,3-diarylsubstituted indole via McMurry coupling for functional characterization of cyclooxygenase-2 (COX-2) in vitro and in vivo. Bioorg Med Chem 2012; 20:3410-21. [PMID: 22560838 DOI: 10.1016/j.bmc.2012.04.022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 04/04/2012] [Accepted: 04/10/2012] [Indexed: 11/18/2022]
Abstract
The radiosynthesis of 3-(4-[(18)F]fluorophenyl)-2-(4-methylsulfonylphenyl)-1H-indole [(18)F]-3 as potential PET radiotracer for functional characterization of cyclooxygenase-2 (COX-2) in vitro and in vivo is described. [(18)F]-3 was prepared by McMurry cyclization of a (18)F-labeled intermediate with low valent titanium and zinc via a two-step procedure in a remote controlled synthesizer unit including HPLC purification and solid phase extraction. In this way [(18)F]-3 was synthesized in 80 min synthesis time in 10% total decay corrected yield from [(18)F]fluoride in radiochemical purity >98% and a specific activity of 74-91 GBq/μmol (EOS). [(18)F]-3 was evaluated in vitro using pro-inflammatory stimulated THP-1 and COX-2 expressing tumor cell lines (FaDu, A2058, HT-29), where the radiotracer uptake was shown to be consistent with up regulated COX-2 expression. The stability of [(18)F]-3 was determined by incubation in rat whole blood and plasma in vitro and by metabolite analysis of arterial blood samples in vivo, showing with 75% of original compound after 60 min an acceptable high metabolic stability. However, no substantial tumor accumulation of [(18)F]-3 could be observed by dynamic small animal PET studies on HT-29 tumor-bearing mice in vivo. This may be due to the only moderate COX-1/COX-2 selectivity of 3 as demonstrated by both cellular and enzymatic cyclooxygenase inhibition assay in vitro. Nevertheless, the new approach first using McMurry cyclization in (18)F-chemistry gives access to (18)F-labeled diarylsubstituted heterocycles that hold promise as radiolabeled COX-2 inhibitors.
Collapse
Affiliation(s)
- Torsten Kniess
- Institute of Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Yaromina A, Krause M, Baumann M. Individualization of cancer treatment from radiotherapy perspective. Mol Oncol 2012; 6:211-21. [PMID: 22381063 DOI: 10.1016/j.molonc.2012.01.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Revised: 01/21/2012] [Accepted: 01/23/2012] [Indexed: 12/25/2022] Open
Abstract
Radiotherapy is today used in about 50% of all cancer patients, often in multidisciplinary approaches. With major advance in radiotherapy techniques, increasing knowledge on tumor genetics and biology and the continuous introduction of specifically targeted drugs into combined radio-oncological treatment schedules, individualization of radiotherapy is of high priority to further improve treatment outcomes, i.e. to increase long-term tumor cure and/or to reduce chronic treatment toxicity. This review gives an overview on the importance of predictive biomarkers for the field of radiation oncology. The current status of knowledge on potential biomarkers of tumor hypoxia, tumor cell metabolism, DNA repair, cancer stem cells and biomarkers for combining radiotherapy with inhibition of the epidermal growth factor receptor using monoclonal antibodies is described.
Collapse
Affiliation(s)
- Ala Yaromina
- Department of Radiation Oncology, OncoRay-National Center for Radiation Research in Oncology, University Hospital and Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Germany
| | | | | |
Collapse
|
26
|
Lamb JM, Robinson C, Bradley J, Laforest R, Dehdashti F, White BM, Wuenschel S, Low DA. Generating lung tumor internal target volumes from 4D-PET maximum intensity projections. Med Phys 2011; 38:5732-7. [PMID: 21992387 DOI: 10.1118/1.3633896] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Positron emission tomography (PET) of lung tumors suffers from breathing-motion induced blurring. Respiratory-correlated PET ameliorates motion blurring and enables visualization of lung tumor functional uptake throughout the breathing cycle but has achieved limited clinical use in radiotherapy planning. In this work, the authors propose a process for generating a gated PET maximum intensity projection (MIP), a breathing-phase projection of the 4D image set comprising gated PET images, as a technique to quantitatively and efficiently incorporate respiratory-correlated PET information into radiotherapy treatment planning. METHODS 4D-CT and respiratory-gated PET using [(18)F]fluorodeoxyglucose (FDG) were acquired of three patients with a total of four small (4-18 cc), clearly defined lower-lobe lung tumors. Internal target volumes (ITVs) for the lung tumors were generated by threshold-based segmentation of PET-MIP images and ungated PET images (ITV(PET-MIP) and ITV(3D-PET), respectively), and by manual contouring of CT-MIP and end-exhale and end-inhale phases of 4D-CT (ITV(CT-MIP)) by a radiation oncologist. Because of the sensitivity of tumor segmentation to threshold value, several different thresholds were tested for ITV generation, including 40%, 30%, and 20% of maximum standardized uptake value (SUV(max)) for FDG as well as absolute SUV thresholds of 2.5 and 3.0. The normalized overlap and relative volumes of ITV(PET-MIP) and ITV(3D-PET) with respect to ITV(CT-MIP) were compared. The images were also visually compared. ITV(CT-MIP) was considered a gold standard for these tumors with CT-visible morphology. RESULTS The mean and standard deviation normalized overlap and relative volumes between ITV(PET-MIP) and ITV(CT-MIP) were 0.68 ± 0.07 and 1.07 ± 0.42, respectively, averaged over all four tumors and all five threshold values. The mean and standard deviation normalized overlap and relative volumes of ITV(3D-PET) and ITV(CT-MIP) were 0.47 ± 0.12 and 0.69 ± 0.56, respectively. CONCLUSIONS PET-MIP images better match CT-MIP images for this sample of four small CT-visible tumors as compared to ungated PET images, based on the metrics of volumetric overlap and relative volumes as well as visual interpretation. The PET-MIP is a way to incorporate 4D-PET imaging into the process of lung tumor contouring that is time-efficient for the radiation oncologist and involves minimal effort to implement in treatment planning software, because it requires only a single PET image beyond contouring on CT alone.
Collapse
Affiliation(s)
- J M Lamb
- Department of Radiation Oncology, University of California, Los Angeles, CA 90095, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Zaleska K, Bruechner K, Baumann M, Zips D, Yaromina A. Tumour-infiltrating CD11b+ myelomonocytes and response to fractionated irradiation of human squamous cell carcinoma (hSCC) xenografts. Radiother Oncol 2011; 101:80-5. [DOI: 10.1016/j.radonc.2011.05.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 05/23/2011] [Accepted: 05/25/2011] [Indexed: 11/27/2022]
|
28
|
Splith K, Bergmann R, Pietzsch J, Neundorf I. Specific targeting of hypoxic tumor tissue with nitroimidazole-peptide conjugates. ChemMedChem 2011; 7:57-61. [PMID: 21956822 DOI: 10.1002/cmdc.201100401] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Indexed: 11/09/2022]
Affiliation(s)
- Katrin Splith
- Institut für Biochemie, Fakultät für Biowissenschaften, Pharmazie und Psychologie, Universität Leipzig, Leipzig, Germany
| | | | | | | |
Collapse
|
29
|
Animal tumor models for PET in drug development. Ann Nucl Med 2011; 25:717-31. [DOI: 10.1007/s12149-011-0531-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 08/16/2011] [Indexed: 10/17/2022]
|
30
|
Aerts HJWL, Bussink J, Oyen WJG, van Elmpt W, Folgering AM, Emans D, Velders M, Lambin P, De Ruysscher D. Identification of residual metabolic-active areas within NSCLC tumours using a pre-radiotherapy FDG-PET-CT scan: a prospective validation. Lung Cancer 2011; 75:73-6. [PMID: 21782272 DOI: 10.1016/j.lungcan.2011.06.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 04/12/2011] [Accepted: 06/09/2011] [Indexed: 11/15/2022]
Abstract
It was recently described that high FDG-uptake areas pre-radiotherapy largely correspond with residual metabolic-active areas post-radiotherapy. Here, an independent prospective validation of these results was performed using an overlap-fraction (OF) calculation of various FDG-uptake based thresholds. Data from twelve patients treated at Radboud University Nijmegen Medical Center with lung cancer were analyzed. All patients underwent two FDG-PET-CT scans, one pre-radiotherapy (pre-RT) and one approximately three months after treatment (post-RT). Of the twelve analyzed patients, eight patients showed residual FDG uptake on the post-RT scan and were included for analysis. One of these patients had a residue that was not clearly distinguishable from the surrounding tissue due to FDG avid inflammation. Therefore, seven patients remained for further analysis. The mean volume of the residual metabolic-active areas post-RT was 14.6±10.0% (mean±SD) of the mean volume of the gross tumour volume (GTV) pre-RT. The residual metabolic-active areas largely corresponded with the pre-RT GTV (OF=93.7±7.2%). The pre-RT-scan threshold delineations of 34%, 40% and 50% of the SUV(max) had a large OF with the residual region, 86.9±8.3%, 77.4±8.1% and 67.9±6.8%, respectively. In this independent dataset, we confirmed that the location of residual FDG-uptake areas after radiotherapy corresponds with the high FDG-uptake areas pre-radiotherapy. Therefore, a pre-radiotherapy FDG-PET-CT scan can potentially be used for radiotherapy dose redistribution.
Collapse
Affiliation(s)
- Hugo J W L Aerts
- Department of Radiation Oncology (MAASTRO), GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Yaromina A, Kroeber T, Meinzer A, Boeke S, Thames H, Baumann M, Zips D. Exploratory Study of the Prognostic Value of Microenvironmental Parameters During Fractionated Irradiation in Human Squamous Cell Carcinoma Xenografts. Int J Radiat Oncol Biol Phys 2011; 80:1205-13. [DOI: 10.1016/j.ijrobp.2011.02.015] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Revised: 01/28/2011] [Accepted: 02/02/2011] [Indexed: 11/29/2022]
|
32
|
Accessing radiation response using hypoxia PET imaging and oxygen sensitive electrodes: a preclinical study. Radiother Oncol 2011; 99:418-23. [PMID: 21723634 DOI: 10.1016/j.radonc.2011.06.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 06/16/2011] [Accepted: 06/16/2011] [Indexed: 12/12/2022]
Abstract
PURPOSE Tumor hypoxia is a known cause of resistance to radiotherapy. The aim of this study was to investigate the prognostic value of hypoxia measured by (18)F-fluoroazomycin arabinoside ((18)F-FAZA) PET or the Eppendorf oxygen electrode in a pre-clinical tumor model. MATERIAL/METHODS Pretreatment (18)F-FAZA PET scans and blood sampling was conducted in 92 Female CDF1 mice with subcutaneous C3H mammary carcinomas grown in the right foot. Similarly, oxygenation status of 80 equivalent tumors was assessed using an invasive oxygen sensitive electrode. Tumors were then irradiated with a single dose of 55 Gy and local tumor control up to 90 days after the treatment was determined. RESULTS A significant difference in local tumor control between "more hypoxic" or "less hypoxic" groups separated either by a median (18)F-FAZA PET determined tumor-to-blood ratio (P=0.007; hazard ratio, HR=0.21 [95% CI: 0.06-0.74]), or the fraction of oxygen partial pressure (pO(2)) values ≤2.5 mmHg (P=0.018; HR=0.31 [95% CI: 0.11-0.87]), was found. Both assays showed that the more hypoxic tumors had significantly lower tumor control. CONCLUSION (18)F-FAZA PET analysis showed that pre treatment tumor hypoxia was prognostic of radiation response. Similar results were obtained when oxygenation status was assessed by the Eppendorf pO(2) Histograph. The results of this study support the role of (18)F-FAZA as a non-invasive prognostic marker for tumor hypoxia.
Collapse
|
33
|
Validation of an amino-acid-based radionuclide therapy plus external beam radiotherapy in heterotopic glioblastoma models. Nucl Med Biol 2011; 38:451-60. [DOI: 10.1016/j.nucmedbio.2010.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2009] [Revised: 11/18/2010] [Accepted: 12/13/2010] [Indexed: 11/19/2022]
|
34
|
van den Bogaard J, Janssen MH, Janssens G, Buijsen J, Reniers B, Lambin P, Lammering G, Öllers MC. Residual metabolic tumor activity after chemo-radiotherapy is mainly located in initially high FDG uptake areas in rectal cancer. Radiother Oncol 2011; 99:137-41. [DOI: 10.1016/j.radonc.2011.04.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 04/06/2011] [Accepted: 04/08/2011] [Indexed: 12/25/2022]
|
35
|
Hentschel M, Appold S, Schreiber A, Abolmaali N, Abramyuk A, Dörr W, Kotzerke J, Baumann M, Zöphel K. Early FDG PET at 10 or 20 Gy under chemoradiotherapy is prognostic for locoregional control and overall survival in patients with head and neck cancer. Eur J Nucl Med Mol Imaging 2011; 38:1203-11. [PMID: 21350962 DOI: 10.1007/s00259-011-1759-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2010] [Accepted: 02/02/2011] [Indexed: 12/13/2022]
Abstract
PURPOSE Our study aimed to explore the optimal timing as well as the most appropriate prognostic parameter of (18)F-fluorodeoxyglucose positron emission tomography (FDG PET) during chemoradiotherapy (CRT) for an early prediction of outcome for patients with head and neck squamous cell carcinoma (HNSCC). METHODS Serial PET data (before and three times during CRT) of 37 patients with advanced stage HNSCC, receiving combined CRT between 2005 and 2009, were evaluated. The maximum standardized uptake value (SUV(max)), the average SUV (SUV(mean)) and the gross tumour volume determined by FDG PET (GTV PET), based on a source to background algorithm, were analysed. Stratified actuarial analysis was performed for overall survival (OS), disease-free survival (DFS) and locoregional control (LRC). The median follow-up time was 26 months (range 8-50). RESULTS For all patients, OS was 51%, DFS 44% and LRC 55% after 2 years. The 2-year OS (88%) and 2-year LRC (88%) were higher for patients whose SUV(max) of the primary tumour decreased 50% or more from the beginning (0 Gy) to week 1 or 2 (10 or 20 Gy) of CRT (ΔSUV(max10/20) ≥ 50%) than for patients with ΔSUV(max20) < 50% (2-year OS = 38%; p = 0.02; 2-year LRC 40%; p = 0.06). A pretreatment GTV PET below the median of 10.2 ml predicted a better 2-year OS (34% for GTV PET ≥ 10.2 ml vs 83% for GTV PET < 10.2 ml; p = 0.02). CONCLUSION The decrease of SUV(max) from before (0 Gy) to week 1 or 2 (10 or 20 Gy) of CRT is a potential prognostic marker for patients with HNSCC. Because GTV PET depends on the applied method of analysis, we suggest the use of SUV(max), especially ΔSUV(max10/20), for an early estimation of therapy outcome. Confirmatory studies are warranted.
Collapse
Affiliation(s)
- Maria Hentschel
- Clinic and Polyclinic of Nuclear Medicine, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Fetscherstraße 74, 01307 Dresden, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
van Elmpt W, Öllers M, van Herwijnen H, den Holder L, Vercoulen L, Wouters M, Lambin P, De Ruysscher D. Volume or Position Changes of Primary Lung Tumor During (Chemo-)Radiotherapy Cannot Be Used as a Surrogate for Mediastinal Lymph Node Changes: The Case for Optimal Mediastinal Lymph Node Imaging During Radiotherapy. Int J Radiat Oncol Biol Phys 2011; 79:89-95. [DOI: 10.1016/j.ijrobp.2009.10.059] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 10/08/2009] [Accepted: 10/23/2009] [Indexed: 11/15/2022]
|
37
|
FDG for dose painting: A rational choice. Radiother Oncol 2010; 97:163-4. [DOI: 10.1016/j.radonc.2010.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 05/16/2010] [Indexed: 12/11/2022]
|
38
|
Lambin P, Petit SF, Aerts HJWL, van Elmpt WJC, Oberije CJG, Starmans MHW, van Stiphout RGPM, van Dongen GAMS, Muylle K, Flamen P, Dekker ALAJ, De Ruysscher D. The ESTRO Breur Lecture 2009. From population to voxel-based radiotherapy: exploiting intra-tumour and intra-organ heterogeneity for advanced treatment of non-small cell lung cancer. Radiother Oncol 2010; 96:145-52. [PMID: 20647155 DOI: 10.1016/j.radonc.2010.07.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Revised: 07/07/2010] [Accepted: 07/07/2010] [Indexed: 01/22/2023]
Abstract
Evidence is accumulating that radiotherapy of non-small cell lung cancer patients can be optimized by escalating the tumour dose until the normal tissue tolerances are met. To further improve the therapeutic ratio between tumour control probability and the risk of normal tissue complications, we firstly need to exploit inter patient variation. This variation arises, e.g. from differences in tumour shape and size, lung function and genetic factors. Secondly improvement is achieved by taking into account intra-tumour and intra-organ heterogeneity derived from molecular and functional imaging. Additional radiation dose must be delivered to those parts of the tumour that need it the most, e.g. because of increased radio-resistance or reduced therapeutic drug uptake, and away from regions inside the lung that are most prone to complication. As the delivery of these treatments plans is very sensitive for geometrical uncertainties, probabilistic treatment planning is needed to generate robust treatment plans. The administration of these complicated dose distributions requires a quality assurance procedure that can evaluate the treatment delivery and, if necessary, adapt the treatment plan during radiotherapy.
Collapse
Affiliation(s)
- Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Firat E, Heinemann F, Grosu AL, Hermann F, Niedermann G. Molecular radiobiology meets clinical radiation oncology. Int J Radiat Biol 2010; 86:252-9. [PMID: 20201653 DOI: 10.3109/09553000903419320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The 2nd Langendorff Congress in Freiburg in Breisgau (Germany) gathered basic and translational scientists as well as clinicians interested in recent developments in molecular and clinical radiobiology. The topics ranged from the most recent insight into the organisation of the DNA damage response and radiotherapeutically relevant cell death mechanisms to biological imaging for treatment planning and advances in the understanding of the molecular biological effects of particle beams. Clinical aspects of stem cell and tumour stem cell biology as well as of angiogenesis and hypoxia, the search for novel molecular radiosensitisers and potential strategies for exploitation of the immune system to further improve tumour radiotherapy were also discussed. RESULTS AND CONCLUSION This report surveys the presentations at the meeting, considering their significance in light of the literature, and documents the increasing importance of molecular radiobiology for clinical radiooncology.
Collapse
Affiliation(s)
- Elke Firat
- Department of Radiation Oncology, University of Freiburg, Freiburg, Germany
| | | | | | | | | |
Collapse
|
40
|
Perrin R, Evans PM, Webb S, Partridge M. The use of PET images for radiotherapy treatment planning: an error analysis using radiobiological endpoints. Med Phys 2010; 37:516-31. [PMID: 20229860 DOI: 10.1118/1.3276776] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2025] Open
Abstract
PURPOSE There is significant current interest in the use of biological image guidance in radiotherapy planning. In lung-cancer treatment, tumor motion due to respiration is known to be a limitation. This is particularly true in PET, where image data are collected over a number of minutes. An in-house-developed 4D PET acquisition mode is described and an analysis of the effects of acquisition parameters on the reconstructed image quality is presented. The potential impact of the resulting biological image quality on radiotherapy planning is then quantified in terms of tumor control probability (TCP). METHODS Data were acquired using a human torso phantom comprised of a hot '8F-filled spheroidal "tumor" (40 mm in diameter) suspended in an air-filled "lung" cylinder and surrounded by a warm 18F-filled background. Two different sphere-to-background (S/B) ratios were used. The tumor was connected to a 3-axis computer-controlled motion stage and could be moved during PET data acquisition. Images were acquired with a range of count statistics, motion blurring, and CT attenuation correction (CTAC) misalignment. Four simple models were proposed for the assignment of clonogenic cell density according to the voxel value. The impact of image artifacts was then assessed by calculating the TCP, which is the probability that no clonogenic tumor cell remains after a given dose of radiation. TCP was calculated for a uniform dose distribution in the tumor. RESULTS Reduced count statistics and misaligned CTAC images had the most detrimental impact on the image fidelity. It was found that in both cases the images became less intense, demonstrated by smaller number of voxels at the maximum values. The maximum TCP difference between images with the least and most noise was 3.4% (S/B=3), and with weakest and strongest CT misalignment artifacts, it was 3.2% (S/B = 10). Motion blurring only contributed weakly to the TCP imprecision at 1.7% (S/B=10) between best- and worst-case images. However, the model-calculated TCP showed increasing differences from the ground truth as the complexity of the model increased [maximum difference of approximately 8% (model 3)], which could be attributed to the partial volume effect. CONCLUSIONS Based on the results of this study, it is believed that simple techniques of biologically guided radiotherapy planning for lung cancer should be feasible at intermediate contrast levels (tumor-to-background ratio of approximately 10) with the clinically achievable image quality.
Collapse
Affiliation(s)
- R Perrin
- Joint Department of Physics, Institute of Cancer Research and Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey SM2 5PT, United Kingdom.
| | | | | | | |
Collapse
|
41
|
Papathanassiou D, Bruna-Muraille C, Liehn JC, Nguyen TD, Curé H. Positron Emission Tomography in oncology: Present and future of PET and PET/CT. Crit Rev Oncol Hematol 2009; 72:239-54. [DOI: 10.1016/j.critrevonc.2008.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 09/30/2008] [Accepted: 10/14/2008] [Indexed: 01/01/2023] Open
|
42
|
Samnick S, Romeike BF, Lehmann T, Israel I, Rübe C, Mautes A, Reiners C, Kirsch CM. Efficacy of Systemic Radionuclide Therapy with p-131I-Iodo-l-Phenylalanine Combined with External Beam Photon Irradiation in Treating Malignant Gliomas. J Nucl Med 2009; 50:2025-32. [DOI: 10.2967/jnumed.109.066548] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
43
|
Grotius J, Dittfeld C, Huether M, Mueller-Klieser W, Baumann M, Kunz-Schughart LA. Impact of exogenous lactate on survival and radioresponse of carcinoma cells in vitro. Int J Radiat Biol 2009; 85:989-1001. [DOI: 10.3109/09553000903242156] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
44
|
Bruechner K, Bergmann R, Santiago A, Mosch B, Yaromina A, Hessel F, Hofheinz F, van den Hoff J, Baumann M, Beuthien-Baumann B. Comparison of [18F]FDG uptake and distribution with hypoxia and proliferation in FaDu human squamous cell carcinoma (hSCC) xenografts after single dose irradiation. Int J Radiat Biol 2009; 85:772-80. [PMID: 19657862 DOI: 10.1080/09553000903043067] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE This study investigated the uptake of [(18)F]2-fluoro-2-deoxy-glucose ([(18)F]FDG) in the human tumour xenograft FaDu at early time points after single dose irradiation with Positron-Emission-Tomography (PET), autoradiography and functional histology. MATERIALS AND METHODS [(18)F]FDG-PET of FaDu hSCC xenografts on nude mice was performed before 25 Gy or 35 Gy single dose irradiation and one, seven or 11 days post irradiation (p.irr.). Before the second PET, mice were injected with pimonidazole (pimo) and bromodeoxyuridine (BrdU). After the PET tumours were excised, sliced and subjected to autoradiography and functional histology staining (pimo, BrdU, Ki67). [(18)F]FDG tumour uptake was quantified in the PET scans by maximal standard uptake value (SUV(max)) and in the autoradiography after co-registration to the histology slices. RESULTS No differences in the overall [(18)F]FDG uptake between the two dose groups and time points were found with PET or autoradiography. Comparing autoradiography and histology, the [(18)F]FDG uptake was constant in tumour necrosis over time, while it decreased in vital tumour areas and particularly in hypoxic regions. No differences in the [(18)F]FDG uptake between positive and negative areas of Ki67 and BrdU were found. CONCLUSIONS The decline of [(18)F]FDG uptake in vital tumour and in pimopositive areas as seen in autoradiography, was not reflected by evaluation of SUV(max) determined by PET. These findings suggest that the SUV(max) does not necessarily reflect changes in tumour biology after irradiation.
Collapse
Affiliation(s)
- Kerstin Bruechner
- Department of Radiation Oncology, Medical Faculty Carl Gustav Carus, University of Technology-Dresden, Fetscherstrasse 74, Dresden,Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Rodemann HP. Molecular radiation biology: Perspectives for radiation oncology. Radiother Oncol 2009; 92:293-8. [PMID: 19726094 DOI: 10.1016/j.radonc.2009.08.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Accepted: 08/15/2009] [Indexed: 12/27/2022]
|
46
|
Metabolic control probability in tumour subvolumes or how to guide tumour dose redistribution in non-small cell lung cancer (NSCLC): An exploratory clinical study. Radiother Oncol 2009; 91:393-8. [DOI: 10.1016/j.radonc.2009.02.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 02/19/2009] [Accepted: 02/27/2009] [Indexed: 11/22/2022]
|
47
|
Abramyuk A, Tokalov S, Zöphel K, Koch A, Szluha Lazanyi K, Gillham C, Herrmann T, Abolmaali N. Is pre-therapeutical FDG-PET/CT capable to detect high risk tumor subvolumes responsible for local failure in non-small cell lung cancer? Radiother Oncol 2009; 91:399-404. [DOI: 10.1016/j.radonc.2009.01.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 01/05/2009] [Indexed: 10/21/2022]
|
48
|
Baumann M, Zips D, Appold S. Radiotherapy of lung cancer: Technology meets biology meets multidisciplinarity. Radiother Oncol 2009; 91:279-81. [DOI: 10.1016/j.radonc.2009.05.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 05/07/2009] [Indexed: 11/26/2022]
|
49
|
Aerts HJWL, van Baardwijk AAW, Petit SF, Offermann C, Loon JV, Houben R, Dingemans AMC, Wanders R, Boersma L, Borger J, Bootsma G, Geraedts W, Pitz C, Simons J, Wouters BG, Oellers M, Lambin P, Bosmans G, Dekker ALAJ, De Ruysscher D. Identification of residual metabolic-active areas within individual NSCLC tumours using a pre-radiotherapy (18)Fluorodeoxyglucose-PET-CT scan. Radiother Oncol 2009; 91:386-92. [PMID: 19329207 DOI: 10.1016/j.radonc.2009.03.006] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 03/02/2009] [Accepted: 03/04/2009] [Indexed: 10/25/2022]
Abstract
BACKGROUND AND PURPOSE Non-small cell lung cancer (NSCLC) tumours are mostly heterogeneous. We hypothesized that areas within the tumour with a high pre-radiation (18)F-deoxyglucose (FDG) uptake, could identify residual metabolic-active areas, ultimately enabling selective-boosting of tumour sub-volumes. MATERIAL AND METHODS Fifty-five patients with inoperable stage I-III NSCLC treated with chemo-radiation or with radiotherapy alone were included. For each patient one pre-radiotherapy and one post-radiotherapy FDG-PET-CT scans were available. Twenty-two patients showing persistent FDG uptake in the primary tumour after radiotherapy were analyzed. Overlap fractions (OFs) were calculated between standardized uptake value (SUV) threshold-based auto-delineations on the pre- and post-radiotherapy scan. RESULTS Patients with residual metabolic-active areas within the tumour had a significantly worse survival compared to individuals with a complete metabolic response (p=0.002). The residual metabolic-active areas within the tumour largely corresponded (OF>70%) with the 50%SUV high FDG uptake area of the pre-radiotherapy scan. The hotspot within the residual area (90%SUV) was completely within the GTV (OF=100%), and had a high overlap with the pre-radiotherapy 50%SUV threshold (OF>84%). CONCLUSIONS The location of residual metabolic-active areas within the primary tumour after therapy corresponded with the original high FDG uptake areas pre-radiotherapy. Therefore, a single pre-treatment FDG-PET-CT scan allows for the identification of residual metabolic-active areas.
Collapse
Affiliation(s)
- Hugo J W L Aerts
- Department of Radiation Oncology (MAASTRO), Grow-School for Oncology and Developmental Biology, Maastricht University, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Quantitative Cell Kill of Radio- and Chemotherapy. THE IMPACT OF TUMOR BIOLOGY ON CANCER TREATMENT AND MULTIDISCIPLINARY STRATEGIES 2009. [DOI: 10.1007/978-3-540-74386-6_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|