1
|
Gui C, Wray R, Schöder H, Deasy JO, Grkovski M, Humm JL, Wong RJ, Sherman EJ, Riaz N, Lee NY. Tumor Hypoxia on 18F-fluoromisonidazole Positron Emission Tomography and Distant Metastasis From Head and Neck Squamous Cell Carcinoma. JAMA Netw Open 2024; 7:e2436407. [PMID: 39348119 PMCID: PMC11443350 DOI: 10.1001/jamanetworkopen.2024.36407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/01/2024] Open
Abstract
Importance Given high rates of locoregional control after definitive management of head and neck squamous cell carcinoma (HNSCC), better methods are needed to project distant metastasis (DM) risk. Tumor hypoxia on 18F-fluoromisonidazole (FMISO) positron emission tomography (PET) is associated with locoregional failure, but data demonstrating an association with DM are limited. Objective To determine whether tumor hypoxia on FMISO PET is associated with DM risk after chemoradiotherapy (CRT) for HNSCC. Design, Setting, and Participants This cohort study assessed patients with HNSCC enrolled in 2 prospective clinical trials at a single academic referral center from 2004 to 2021 in which participants received FMISO PET before and during CRT. Data analysis occurred from May 2023 to May 2024. Exposures FMISO PET scans before and 1 to 2 weeks after starting CRT were evaluated for tumor hypoxia by nuclear medicine physicians. Main Outcomes and Measures The primary outcome was DM, defined as biopsy-proven HNSCC outside the primary site and regional lymph nodes. Time to DM was modeled with competing risk regression, with death as a competing risk. Overall survival (OS) was assessed secondarily and modeled with Cox regression. Results Among 281 patients (median [range] age at CRT, 58.7 [25.5-85.6] years; 251 male [89.3%]) included in this study, 242 (86.1%) had oropharyngeal primary cancer, and 266 (94.7%) had human papillomavirus-positive disease. Of all patients, 217 (77.2%) had T stage 1 or 2, and 231 patients (82.2%) had N stage 2b or less. De-escalated 30 Gy CRT was delivered to 144 patients (51.2%), and the remainder received standard 70 Gy CRT. On FMISO PET examination, 73 patients (26.0%) had hypoxia-negative disease before CRT, 138 patients (49.1%) had hypoxia-positive disease before CRT and then hypoxia-negative disease during CRT, and 70 patients (24.9%) persistently had hypoxia-positive disease before and during CRT. At a median (IQR) 58 (46-91) months of follow-up, 12 DM events and 22 deaths were observed. Persistent intratreatment hypoxia was associated with increased DM risk (hazard ratio, 3.51; 95% CI, 1.05-11.79; P = .04) and worse OS (hazard ratio, 2.66; 95% CI, 1.14-6.19; P = .02). No patients with hypoxia-negative disease before CRT experienced DM. Conclusions and Relevance In this cohort study using pooled analysis of prospective nonrandomized clinical trials incorporating FMISO PET in the definitive management of HNSCC, persistent intratreatment hypoxia was associated with increased risk of DM and worse OS. Conversely, all patients with hypoxia-negative disease before treatment remained free of DM. These findings suggest that pretreatment and intratreatment FMISO PET results may serve as biomarkers for DM risk and aid in identifying candidates for escalated therapeutic strategies.
Collapse
Affiliation(s)
- Chengcheng Gui
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rick Wray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Joseph O Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John L Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Richard J Wong
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Immunogenomics and Precision Oncology Platform, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
2
|
Caldarella C, De Risi M, Massaccesi M, Miccichè F, Bussu F, Galli J, Rufini V, Leccisotti L. Role of 18F-FDG PET/CT in Head and Neck Squamous Cell Carcinoma: Current Evidence and Innovative Applications. Cancers (Basel) 2024; 16:1905. [PMID: 38791983 PMCID: PMC11119768 DOI: 10.3390/cancers16101905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
This article provides an overview of the use of 18F-FDG PET/CT in various clinical scenarios of head-neck squamous cell carcinoma, ranging from initial staging to treatment-response assessment, and post-therapy follow-up, with a focus on the current evidence, debated issues, and innovative applications. Methodological aspects and the most frequent pitfalls in head-neck imaging interpretation are described. In the initial work-up, 18F-FDG PET/CT is recommended in patients with metastatic cervical lymphadenectomy and occult primary tumor; moreover, it is a well-established imaging tool for detecting cervical nodal involvement, distant metastases, and synchronous primary tumors. Various 18F-FDG pre-treatment parameters show prognostic value in terms of disease progression and overall survival. In this scenario, an emerging role is played by radiomics and machine learning. For radiation-treatment planning, 18F-FDG PET/CT provides an accurate delineation of target volumes and treatment adaptation. Due to its high negative predictive value, 18F-FDG PET/CT, performed at least 12 weeks after the completion of chemoradiotherapy, can prevent unnecessary neck dissections. In addition to radiomics and machine learning, emerging applications include PET/MRI, which combines the high soft-tissue contrast of MRI with the metabolic information of PET, and the use of PET radiopharmaceuticals other than 18F-FDG, which can answer specific clinical needs.
Collapse
Affiliation(s)
- Carmelo Caldarella
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
| | - Marina De Risi
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
| | - Mariangela Massaccesi
- Radiation Oncology Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Francesco Miccichè
- Radiation Oncology Unit, Ospedale Isola Tiberina—Gemelli Isola, 00186 Rome, Italy;
| | - Francesco Bussu
- Otorhinolaryngology Operative Unit, Azienda Ospedaliero Universitaria Sassari, 07100 Sassari, Italy;
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Jacopo Galli
- Otorhinolaryngology Unit, Department of Neurosciences, Sensory Organs and Thorax, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Section of Otolaryngology, Department of Head-Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Vittoria Rufini
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
- Section of Nuclear Medicine, Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lucia Leccisotti
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
- Section of Nuclear Medicine, Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
3
|
Henjum H, Dahle TJ, Mairani A, Pilskog S, Stokkevåg C, Boer CG, Redalen KR, Minn H, Malinen E, Ytre‐Hauge KS. Combined RBE and OER optimization in proton therapy with FLUKA based on EF5-PET. J Appl Clin Med Phys 2023; 24:e14014. [PMID: 37161820 PMCID: PMC10476997 DOI: 10.1002/acm2.14014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/14/2023] [Accepted: 04/10/2023] [Indexed: 05/11/2023] Open
Abstract
INTRODUCTION Tumor hypoxia is associated with poor treatment outcome. Hypoxic regions are more radioresistant than well-oxygenated regions, as quantified by the oxygen enhancement ratio (OER). In optimization of proton therapy, including OER in addition to the relative biological effectiveness (RBE) could therefore be used to adapt to patient-specific radioresistance governed by intrinsic radiosensitivity and hypoxia. METHODS A combined RBE and OER weighted dose (ROWD) calculation method was implemented in a FLUKA Monte Carlo (MC) based treatment planning tool. The method is based on the linear quadratic model, with α and β parameters as a function of the OER, and therefore a function of the linear energy transfer (LET) and partial oxygen pressure (pO2 ). Proton therapy plans for two head and neck cancer (HNC) patients were optimized with pO2 estimated from [18 F]-EF5 positron emission tomography (PET) images. For the ROWD calculations, an RBE of 1.1 (RBE1.1,OER ) and two variable RBE models, Rørvik (ROR) and McNamara (MCN), were used, alongside a reference plan without incorporation of OER (RBE1.1 ). RESULTS For the HNC patients, treatment plans in line with the prescription dose and with acceptable target ROWD could be generated with the established tool. The physical dose was the main factor modulated in the ROWD. The impact of incorporating OER during optimization of HNC patients was demonstrated by the substantial difference found between ROWD and physical dose in the hypoxic tumor region. The largest physical dose differences between the ROWD optimized plans and the reference plan was 12.2 Gy. CONCLUSION The FLUKA MC based tool was able to optimize proton treatment plans taking the tumor pO2 distribution from hypoxia PET images into account. Independent of RBE-model, both elevated LET and physical dose were found in the hypoxic regions, which shows the potential to increase the tumor control compared to a conventional optimization approach.
Collapse
Affiliation(s)
- Helge Henjum
- Department of Physics and TechnologyUniversity of BergenBergenNorway
| | - Tordis Johnsen Dahle
- Department of Physics and TechnologyUniversity of BergenBergenNorway
- Department of Oncology and Medical PhysicsHaukeland University HospitalBergenNorway
| | - Andrea Mairani
- Centro Nazionale di Adroterapia Oncologica (CNAO Foundation)PaviaItaly
- Heidelberg Ion Beam Therapy Center (HIT)HeidelbergGermany
| | - Sara Pilskog
- Department of Physics and TechnologyUniversity of BergenBergenNorway
- Department of Oncology and Medical PhysicsHaukeland University HospitalBergenNorway
| | - Camilla Stokkevåg
- Department of Physics and TechnologyUniversity of BergenBergenNorway
- Department of Oncology and Medical PhysicsHaukeland University HospitalBergenNorway
| | | | - Kathrine Røe Redalen
- Department of PhysicsNorwegian University of Science and TechnologyTrondheimNorway
| | - Heikki Minn
- Department of Oncology and RadiotherapyTurku University HospitalTurkuFinland
- Turku PET CentreUniversity of TurkuTurkuFinland
| | - Eirik Malinen
- Department of PhysicsUniversity of OsloOsloNorway
- Department of Medical PhysicsOslo University HospitalOsloNorway
| | | |
Collapse
|
4
|
Boeke S, Winter RM, Leibfarth S, Krueger MA, Bowden G, Cotton J, Pichler BJ, Zips D, Thorwarth D. Machine learning identifies multi-parametric functional PET/MR imaging cluster to predict radiation resistance in preclinical head and neck cancer models. Eur J Nucl Med Mol Imaging 2023; 50:3084-3096. [PMID: 37148296 PMCID: PMC10382355 DOI: 10.1007/s00259-023-06254-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
PURPOSE Tumor hypoxia and other microenvironmental factors are key determinants of treatment resistance. Hypoxia positron emission tomography (PET) and functional magnetic resonance imaging (MRI) are established prognostic imaging modalities to identify radiation resistance in head-and-neck cancer (HNC). The aim of this preclinical study was to develop a multi-parametric imaging parameter specifically for focal radiotherapy (RT) dose escalation using HNC xenografts of different radiation sensitivities. METHODS A total of eight human HNC xenograft models were implanted into 68 immunodeficient mice. Combined PET/MRI using dynamic [18F]-fluoromisonidazole (FMISO) hypoxia PET, diffusion-weighted (DW), and dynamic contrast-enhanced MRI was carried out before and after fractionated RT (10 × 2 Gy). Imaging data were analyzed on voxel-basis using principal component (PC) analysis for dynamic data and apparent diffusion coefficients (ADCs) for DW-MRI. A data- and hypothesis-driven machine learning model was trained to identify clusters of high-risk subvolumes (HRSs) from multi-dimensional (1-5D) pre-clinical imaging data before and after RT. The stratification potential of each 1D to 5D model with respect to radiation sensitivity was evaluated using Cohen's d-score and compared to classical features such as mean/peak/maximum standardized uptake values (SUVmean/peak/max) and tumor-to-muscle-ratios (TMRpeak/max) as well as minimum/valley/maximum/mean ADC. RESULTS Complete 5D imaging data were available for 42 animals. The final preclinical model for HRS identification at baseline yielding the highest stratification potential was defined in 3D imaging space based on ADC and two FMISO PCs ([Formula: see text]). In 1D imaging space, only clusters of ADC revealed significant stratification potential ([Formula: see text]). Among all classical features, only ADCvalley showed significant correlation to radiation resistance ([Formula: see text]). After 2 weeks of RT, FMISO_c1 showed significant correlation to radiation resistance ([Formula: see text]). CONCLUSION A quantitative imaging metric was described in a preclinical study indicating that radiation-resistant subvolumes in HNC may be detected by clusters of ADC and FMISO using combined PET/MRI which are potential targets for future functional image-guided RT dose-painting approaches and require clinical validation.
Collapse
Affiliation(s)
- Simon Boeke
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - René M Winter
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Sara Leibfarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Marcel A Krueger
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Gregory Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Jonathan Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| |
Collapse
|
5
|
Abstract
Hypoxia (oxygen deprivation) occurs in most solid malignancies, albeit with considerable heterogeneity. Hypoxia is associated with an aggressive cancer phenotype by promotion of genomic instability, evasion of anti-cancer therapies including radiotherapy and enhancement of metastatic risk. Therefore, hypoxia results in poor cancer outcomes. Targeting hypoxia to improve cancer outcomes is an attractive therapeutic strategy. Hypoxia-targeted dose painting escalates radiotherapy dose to hypoxic sub-volumes, as quantified and spatially mapped using hypoxia imaging. This therapeutic approach could overcome hypoxia-induced radioresistance and improve patient outcomes without the need for hypoxia-targeted drugs. This article will review the premise and underpinning evidence for personalized hypoxia-targeted dose painting. It will present data on relevant hypoxia imaging biomarkers, highlight the challenges and potential benefit of this approach and provide recommendations for future research priorities in this field. Personalized hypoxia-based radiotherapy de-escalation strategies will also be addressed.
Collapse
Affiliation(s)
- Ahmed Salem
- Department of Anatomy, Physiology and Biochemistry, Faculty of Medicine, Hashemite University, Zarqa, Jordan; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
6
|
Sommat K, Tong AKT, Ong ALK, Hu J, Sin SY, Lam WWC, Xie W, Khor YM, Lim C, Lim TW, Selvarajan S, Wang F, Tan TWK, Wee JTS, Soong YL, Fong KW, Hennedige T, Hua TC. 18F-FMISO PET-guided dose escalation with multifield optimization intensity-modulated proton therapy in nasopharyngeal carcinoma. Asia Pac J Clin Oncol 2023. [PMID: 37157884 DOI: 10.1111/ajco.13953] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/13/2023] [Accepted: 03/22/2023] [Indexed: 05/10/2023]
Abstract
PURPOSE The purpose of this study was to evaluate the radiotherapy planning feasibility of dose escalation with intensity-modulated proton therapy (IMPT) to hypoxic tumor regions identified on 18F-Fluoromisonidazole (FMISO) positron emission tomography and computed tomography (PET-CT) in NPC. MATERIALS AND METHODS Nine patients with stages T3-4N0-3M0 NPC underwent 18F-FMISO PET-CT before and during week 3 of radiotherapy. The hypoxic volume (GTVhypo) is automatically generated by applying a subthresholding algorithm within the gross tumor volume (GTV) with a tumor to muscle standardized uptake value (SUV) ratio of 1.3 on the 18F-FMISO PET-CT scan. Two proton plans were generated for each patient, a standard plan to 70 Gy and dose escalation plan with upfront boost followed by standard 70GyE plan. The stereotactic boost was planned with single-field uniform dose optimization using two fields to deliver 10 GyE in two fractions to GTVhypo. The standard plan was generated with IMPT with robust optimization to deliver 70GyE, 60GyE in 33 fractions using simultaneous integrated boost technique. A plan sum was generated for assessment. RESULTS Eight of nine patients showed tumor hypoxia on the baseline 18F-FMISO PET-CT scan. The mean hypoxic tumor volume was 3.9 cm3 (range .9-11.9cm3 ). The average SUVmax of the hypoxic volume was 2.2 (range 1.44-2.98). All the dose-volume parameters met the planning objectives for target coverage. Dose escalation was not feasible in three of eight patients as the D0.03cc of temporal lobe was greater than 75GyE. CONCLUSIONS The utility of boost to the hypoxic volume before standard course of radiotherapy with IMPT is dosimetrically feasible in selected patients. Clinical trials are warranted to determine the clinical outcomes of this approach.
Collapse
Affiliation(s)
- Kiattisa Sommat
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Aaron Kian Ti Tong
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Ashley Li Kuan Ong
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Jing Hu
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Sze Yarn Sin
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Winnie Wing Chuen Lam
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Wanying Xie
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Yiu Ming Khor
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Cindy Lim
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Tze Wei Lim
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Sathiyamoorthy Selvarajan
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Fuqiang Wang
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Terence Wee Kiat Tan
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Joseph Tien Seng Wee
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Yoke Lim Soong
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Kam Weng Fong
- Division of Radiation Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Tiffany Hennedige
- Division of Oncologic Imaging, National Cancer Centre Singapore, Singapore, Singapore
| | - Thng Choon Hua
- Division of Oncologic Imaging, National Cancer Centre Singapore, Singapore, Singapore
| |
Collapse
|
7
|
Embring A, Onjukka E, Mercke C, Lax I, Berglund A, Friesland S. Dose Escalation of Oropharyngeal Cancer: Long-Time Follow-Up and Side Effects. Cancers (Basel) 2023; 15:cancers15092580. [PMID: 37174046 PMCID: PMC10177133 DOI: 10.3390/cancers15092580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/13/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Previous studies on dose-escalated radiotherapy in head and neck cancer have shown mixed results, and it is not established which patients would benefit from dose escalation. Further, while dose escalation does not appear to increase late toxicity, this needs to be confirmed with longer follow-up. In this study, we analysed treatment outcome and toxicity in 215 patients with oropharyngeal cancer treated with dose-escalated radiotherapy (>72 Gy, EQD2, α/β = 10 Gy, boost by brachytherapy or simultaneous integrated boost) and a matched cohort of 215 patients treated with standard dose external-beam radiotherapy (68 Gy) between 2011 and 2018 at our institution. The 5-year overall survival (OS) was 77.8% (72.4-83.6) and 73.7% (67.8-80.1) in the dose-escalated and standard dose group, respectively (p = 0.24). Median follow-up was 78.1 (49.2-98.4) and 60.2 (38.9-89.4) months in the dose-escalated and standard dose groups, respectively. Grade ≥3 osteoradionecrosis (ORN) and late dysphagia were more common in the dose-escalated group compared to the standard dose group, with 19 (8.8%) vs. 4 (1.9%) patients developing grade ≥3 ORN (p = 0.001), and 39 (18.1%) vs. 21 (9.8%) patients developing grade ≥3 dysphagia (p = 0.01). No predictive factors to help select patients for dose-escalated radiotherapy were found. However, the remarkably good OS in the dose-escalated cohort, despite a predominance of advanced tumour stages, encourages further attempts to identify such factors.
Collapse
Affiliation(s)
- Anna Embring
- Department of Oncology, Karolinska University Hospital, 17176 Stockholm, Sweden
- Karolinska Institute, Department of Oncology-Pathology, 17176 Stockholm, Sweden
| | - Eva Onjukka
- Karolinska Institute, Department of Oncology-Pathology, 17176 Stockholm, Sweden
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Claes Mercke
- Department of Oncology, Karolinska University Hospital, 17176 Stockholm, Sweden
- Karolinska Institute, Department of Oncology-Pathology, 17176 Stockholm, Sweden
| | - Ingmar Lax
- Karolinska Institute, Department of Oncology-Pathology, 17176 Stockholm, Sweden
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Anders Berglund
- Epistat Epidemiology and Statistics Consulting, 75655 Uppsala, Sweden
| | - Signe Friesland
- Department of Oncology, Karolinska University Hospital, 17176 Stockholm, Sweden
- Karolinska Institute, Department of Oncology-Pathology, 17176 Stockholm, Sweden
| |
Collapse
|
8
|
Embring A, Onjukka E, Mercke C, Lax I, Berglund A, Friesland S. Dose escalation in oropharyngeal cancer: a comparison of simultaneous integrated boost and brachytherapy boost. Radiat Oncol 2023; 18:65. [PMID: 37029424 PMCID: PMC10082532 DOI: 10.1186/s13014-023-02256-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Local recurrence is the most common pattern of failure in head and neck cancer. It can therefore be hypothesised that some of these patients would benefit from an intensified local treatment, such as radiation dose escalation of the primary tumour. This study compares treatment and toxicity outcomes from two different boost modalities in oropharyngeal cancer: simultaneous integrated boost (SIB) and brachytherapy boost. METHODS Two hundred and forty-four consecutive patients treated with > 72 Gy for oropharyngeal squamous cell carcinoma between 2011 and 2018 at our institution were retrospectively analysed. Data on side effects were collected from a local quality registry and supplemented with a review of medical records. Patients receiving a brachytherapy boost first had external beam radiotherapy consisting of 68 Gy in 2 Gy fractions to the gross tumour volume (GTV), and elective radiotherapy to the neck bilaterally. The brachytherapy boost was typically given using pulsed dose rate, 15 fractions and 0.56-0.66 Gy per fraction [total dose in EQD2 = 75.4-76.8 Gy (α/β = 10)]. The typical dose escalated radiotherapy with external beam radiotherapy only, was delivered using SIB with 74,8 Gy in 2.2 Gy fractions [EQD2 = 76.0 Gy (α/β = 10)] to the primary tumour, 68 Gy in 2 Gy fractions to GTV + 10 mm margin and elective radiotherapy to the neck bilaterally. RESULTS Dose escalation by SIB was given to 111 patients and brachytherapy boost to 134 patients. The most common type of cancer was base of tongue (55%), followed by tonsillar cancer (42%). The majority of patients had T3- or T4-tumours and 84% were HPV-positive. The 5-year OS was 72,4% (95% CI 66.9-78.3) and the median follow-up was 6.1 years. Comparing the two different dose escalation modalities we found no significant differences in OS or PFS and these results remained after a propensity-score matched analysis was performed. The analysis of grade ≥ 3 side effects showed no significant differences between the two different dose escalation techniques. CONCLUSIONS We found no significant differences in survival or grade ≥ 3 side effects comparing simultaneous integrated boost and brachytherapy boost as alternative dose escalation modalities in the treatment of oropharyngeal cancer.
Collapse
Affiliation(s)
- Anna Embring
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Oncology, Karolinska University Hospital, Anna Steckséns Gata 41, 171 76, Solna, Stockholm, Sweden.
| | - Eva Onjukka
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Claes Mercke
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Karolinska University Hospital, Anna Steckséns Gata 41, 171 76, Solna, Stockholm, Sweden
| | - Ingmar Lax
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Berglund
- Epistat Epidemiology and Statistics Consulting, Uppsala, Sweden
| | - Signe Friesland
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology, Karolinska University Hospital, Anna Steckséns Gata 41, 171 76, Solna, Stockholm, Sweden
| |
Collapse
|
9
|
Thorwarth D. Clinical use of positron emission tomography for radiotherapy planning - Medical physics considerations. Z Med Phys 2023; 33:13-21. [PMID: 36272949 PMCID: PMC10068574 DOI: 10.1016/j.zemedi.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/17/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022]
Abstract
PET/CT imaging plays an increasing role in radiotherapy treatment planning. The aim of this article was to identify the major use cases and technical as well as medical physics challenges during integration of these data into treatment planning. Dedicated aspects, such as (i) PET/CT-based radiotherapy simulation, (ii) PET-based target volume delineation, (iii) functional avoidance to optimized organ-at-risk sparing and (iv) functionally adapted individualized radiotherapy are discussed in this article. Furthermore, medical physics aspects to be taken into account are summarized and presented in form of check-lists.
Collapse
Affiliation(s)
- Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
10
|
Sminia P, Guipaud O, Viktorsson K, Ahire V, Baatout S, Boterberg T, Cizkova J, Dostál M, Fernandez-Palomo C, Filipova A, François A, Geiger M, Hunter A, Jassim H, Edin NFJ, Jordan K, Koniarová I, Selvaraj VK, Meade AD, Milliat F, Montoro A, Politis C, Savu D, Sémont A, Tichy A, Válek V, Vogin G. Clinical Radiobiology for Radiation Oncology. RADIOBIOLOGY TEXTBOOK 2023:237-309. [DOI: 10.1007/978-3-031-18810-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
AbstractThis chapter is focused on radiobiological aspects at the molecular, cellular, and tissue level which are relevant for the clinical use of ionizing radiation (IR) in cancer therapy. For radiation oncology, it is critical to find a balance, i.e., the therapeutic window, between the probability of tumor control and the probability of side effects caused by radiation injury to the healthy tissues and organs. An overview is given about modern precision radiotherapy (RT) techniques, which allow optimal sparing of healthy tissues. Biological factors determining the width of the therapeutic window are explained. The role of the six typical radiobiological phenomena determining the response of both malignant and normal tissues in the clinic, the 6R’s, which are Reoxygenation, Redistribution, Repopulation, Repair, Radiosensitivity, and Reactivation of the immune system, is discussed. Information is provided on tumor characteristics, for example, tumor type, growth kinetics, hypoxia, aberrant molecular signaling pathways, cancer stem cells and their impact on the response to RT. The role of the tumor microenvironment and microbiota is described and the effects of radiation on the immune system including the abscopal effect phenomenon are outlined. A summary is given on tumor diagnosis, response prediction via biomarkers, genetics, and radiomics, and ways to selectively enhance the RT response in tumors. Furthermore, we describe acute and late normal tissue reactions following exposure to radiation: cellular aspects, tissue kinetics, latency periods, permanent or transient injury, and histopathology. Details are also given on the differential effect on tumor and late responding healthy tissues following fractionated and low dose rate irradiation as well as the effect of whole-body exposure.
Collapse
|
11
|
Hildingsson S, Gebre-Medhin M, Zschaeck S, Adrian G. Hypoxia in relationship to tumor volume using hypoxia PET-imaging in head & neck cancer - A scoping review. Clin Transl Radiat Oncol 2022; 36:40-46. [PMID: 35769424 PMCID: PMC9234341 DOI: 10.1016/j.ctro.2022.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023] Open
Abstract
Primary tumor volume and hypoxic volume has previously not been convincingly related. 367 patients with head and neck squamous cell carcinoma from 21 different studies using hypoxia-PET The hypoxic volume increased significantly with primary tumor volume. In larger tumor the hypoxic fraction was significantly higher than in smaller tumors.
Background Hypoxia and large tumor volumes are negative prognostic factors for patients with head and neck squamous cell carcinoma (HNSCC) treated with radiation therapy (RT). PET-scanning with specific hypoxia-tracers (hypoxia-PET) can be used to non-invasively assess hypoxic tumor volume. Primary tumor volume is readily available for patients undergoing RT. However, the relationship between hypoxic volume and primary tumor volume is yet an open question. The current study investigates the hypotheses that larger tumors contain both a larger hypoxic volume and a higher hypoxic fraction. Methods PubMed and Embase were systematically searched to identify articles fulfilling the predefined criteria. Individual tumor data (primary tumor volume and hypoxic volume/fraction) was extracted. Relationship between hypoxic volume and primary tumor volume was investigated by linear regression. The correlation between hypoxic fraction and log2(primary tumor volume) was determined for each cohort and in a pooled analysis individual regression slopes and coefficients of determination (R2) were weighted according to cohort size. Results 21 relevant articles were identified and individual data from 367 patients was extracted, out of which 323 patients from 17 studies had quantifiable volumes of interest. A correlation between primary tumor volume and PET-determined hypoxic volume was found (P <.001, R2 = 0.46). Larger tumors had a significantly higher fraction of hypoxia compared with smaller tumors (P<.01). The weighted analysis of all studies revealed that for each doubling of the tumor volume, the hypoxic fraction increased by four percentage points. Conclusion This study shows correlations between primary tumor volume and hypoxic volume as well as primary tumor volume and the hypoxic fraction in patients with HNSCC. The findings suggest that not only do large tumors contain more cancer cells, they also have a higher proportion of potentially radioresistant hypoxic cells. This knowledge can be important when individualizing RT.
Collapse
Affiliation(s)
- Sofia Hildingsson
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Gebre-Medhin
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gabriel Adrian
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Li G, Wu X, Ma X. Artificial intelligence in radiotherapy. Semin Cancer Biol 2022; 86:160-171. [PMID: 35998809 DOI: 10.1016/j.semcancer.2022.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/18/2022] [Indexed: 11/19/2022]
Abstract
Radiotherapy is a discipline closely integrated with computer science. Artificial intelligence (AI) has developed rapidly over the past few years. With the explosive growth of medical big data, AI promises to revolutionize the field of radiotherapy through highly automated workflow, enhanced quality assurance, improved regional balances of expert experiences, and individualized treatment guided by multi-omics. In addition to independent researchers, the increasing number of large databases, biobanks, and open challenges significantly facilitated AI studies on radiation oncology. This article reviews the latest research, clinical applications, and challenges of AI in each part of radiotherapy including image processing, contouring, planning, quality assurance, motion management, and outcome prediction. By summarizing cutting-edge findings and challenges, we aim to inspire researchers to explore more future possibilities and accelerate the arrival of AI radiotherapy.
Collapse
Affiliation(s)
- Guangqi Li
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xin Wu
- Head & Neck Oncology ward, Division of Radiotherapy Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China
| | - Xuelei Ma
- Division of Biotherapy, Cancer Center, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, No. 37 GuoXue Alley, Chengdu 610041, China.
| |
Collapse
|
13
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
14
|
Adrian G, Carlsson H, Kjellén E, Sjövall J, Zackrisson B, Nilsson P, Gebre-Medhin M. Primary tumor volume and prognosis for patients with p16-positive and p16-negative oropharyngeal squamous cell carcinoma treated with radiation therapy. Radiat Oncol 2022; 17:107. [PMID: 35701827 PMCID: PMC9195357 DOI: 10.1186/s13014-022-02074-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The prescribed radiation dose to patients with oropharyngeal squamous cell carcinoma (OPSCC) is standardized, even if the prognosis for individual patients may differ. Easy-at-hand pre-treatment risk stratification methods are valuable to individualize therapy. In the current study we assessed the prognostic impact of primary tumor volume for p16-positive and p16-negative tumors and in relationship to other prognostic factors for outcome in patients with OPSCC treated with primary radiation therapy (RT). METHODS Five hundred twenty-three OPSCC patients with p16-status treated with primary RT (68.0 Gy to 73.1 Gy in 7 weeks, or 68.0 Gy in 4.5 weeks), with or without concurrent chemotherapy, within three prospective trials were included in the study. Local failure (LF), progression free survival (PFS) and overall survival (OS) in relationship to the size of the primary gross tumor volume (GTV-T) and other prognostic factors were investigated. Efficiency of intensified RT (RT with total dose 73.1 Gy or given within 4.5 weeks) was analyzed in relationship to tumor volume. RESULTS The volume of GTV-T and p16-status were found to be the strongest prognostic markers for LF, PFS and OS. For p16-positive tumors, an increase in tumor volume had a significantly higher negative prognostic impact compared with p16-negative tumors. Within a T-classification, patients with a smaller tumor, compared with a larger tumor, had a better prognosis. The importance of tumor volume remained after adjusting for nodal status, age, performance status, smoking status, sex, and hemoglobin-level. The adjusted hazard ratio for OS per cm3 increase in tumor volume was 2.3% (95% CI 0-4.9) for p16-positive and 1.3% (95% 0.3-2.2) for p16-negative. Exploratory analyses suggested that intensified RT could mitigate the negative impact of a large tumor volume. CONCLUSIONS Outcome for patients with OPSCC treated with RT is largely determined by tumor volume, even when adjusting for other established prognostic factors. Tumor volume is significantly more influential for patients with p16-positive tumors. Patients with large tumor volumes might benefit by intensified RT to improve survival.
Collapse
Affiliation(s)
- Gabriel Adrian
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, Lund, Sweden
| | - Henrik Carlsson
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
- Division of Oncology, Department of Clinical Sciences Lund, Skåne University Hospital, Lund University, Lund, Sweden
| | - Elisabeth Kjellén
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Johanna Sjövall
- Department of Otorhinolaryngology –Head and Neck Surgery, Skåne University Hospital, Lund University, Lund, Sweden
| | - Björn Zackrisson
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Per Nilsson
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences, Medical Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Maria Gebre-Medhin
- Department of Hematology, Oncology, and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
15
|
D’Urso P, Farneti A, Marucci L, Marzi S, Piludu F, Vidiri A, Sanguineti G. Predictors of Outcome after (Chemo)Radiotherapy for Node-Positive Oropharyngeal Cancer: The Role of Functional MRI. Cancers (Basel) 2022; 14:cancers14102477. [PMID: 35626084 PMCID: PMC9139324 DOI: 10.3390/cancers14102477] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/04/2023] Open
Abstract
The prognosis of a subset of patients with locally advanced oropharyngeal cancer (LA-OPC) is still poor despite improvements in patient selection and treatment. Identifying specific patient- and tumor-related factors can help to select those patients who need intensified treatment. We aimed to assess the role of historical risk factors and novel magnetic resonance imaging (MRI) biomarkers in predicting outcomes in these patients. Patients diagnosed with LA-OPC were studied with diffusion-weighted imaging (DWI) and dynamic-contrast enhanced MRI at baseline and at the 10th radiotherapy (RT) fraction. Clinical information was collected as well. The endpoint of the study was the development of disease progression, locally or distantly. Of the 97 patients enrolled, 68 were eligible for analysis. Disease progression was recorded in 21 patients (11 had loco-regional progression, 10 developed distant metastases). We found a correlation between N diameter and disease control (p = 0.02); features such as p16 status and extranodal extension only showed a trend towards statistical significance. Among perfusion MRI features, higher median values of Kep both in primary tumor (T, p = 0.016) and lymph node (N, p = 0.003) and lower median values of ve (p = 0.018 in T, p = 0.004 in N) correlated with better disease control. Kep P90 and N diameter were identified by MRMR algorithm as the best predictors of outcome. In conclusion, the association of non-invasive MRI biomarkers and patients and tumor characteristics may help in predicting disease behavior and patient outcomes in order to ensure a more customized treatment.
Collapse
Affiliation(s)
- Pasqualina D’Urso
- Department of Radiotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.F.); (L.M.); (G.S.)
- Correspondence:
| | - Alessia Farneti
- Department of Radiotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.F.); (L.M.); (G.S.)
| | - Laura Marucci
- Department of Radiotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.F.); (L.M.); (G.S.)
| | - Simona Marzi
- Medical Physics Laboratory, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Francesca Piludu
- Radiology and Diagnostic Imaging Department, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (F.P.); (A.V.)
| | - Antonello Vidiri
- Radiology and Diagnostic Imaging Department, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (F.P.); (A.V.)
| | - Giuseppe Sanguineti
- Department of Radiotherapy, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy; (A.F.); (L.M.); (G.S.)
| |
Collapse
|
16
|
Welz S, Paulsen F, Pfannenberg C, Reimold M, Reischl G, Nikolaou K, La Fougère C, Alber M, Belka C, Zips D, Thorwarth D. Dose escalation to hypoxic subvolumes in head and neck cancer: A randomized phase II study using dynamic [ 18F]FMISO PET/CT. Radiother Oncol 2022; 171:30-36. [PMID: 35395276 DOI: 10.1016/j.radonc.2022.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Tumor hypoxia is a major cause of resistance to radiochemotherapy in locally advanced head-and-neck cancer (LASCCHN). We present results of a randomized phase II trial on hypoxia dose escalation (DE) in LASCCHN based on dynamic [18F]FMISO (dynFMISO) positron emission tomography (PET). The purpose was to confirm the prognostic value of hypoxia PET and assess feasibility, toxicity and efficacy of hypoxia-DE. MATERIALS AND METHODS Patients with LASCCHN underwent baseline dynFMISO PET/CT. Hypoxic volumes (HV) were derived from dynFMISO data. Patients with hypoxic tumors (HV>0) were randomized into standard radiotherapy (ST: 70Gy/35fx) or dose escalation (DE: 77Gy/35fx) to the HV. Patients with non-hypoxic tumors were treated with ST. After a minimum follow-up of 2 years, feasibility, acute/late toxicity and local control (LC) were analyzed. RESULTS The study was closed prematurely due to slow accrual. Between 2009 and 2017, 53 patients were enrolled, 39 (74%) had hypoxic tumors and were randomized into ST or DE. For non-hypoxic patients, 100% 5-year LC was observed compared to 74% in patients with hypoxic tumors (p=0.039). The difference in 5-year LC between DE (16/19) and ST (10/17) was 25%, p=0.150. No relevant differences related to acute and late toxicities between the groups were observed. CONCLUSION This study confirmed the prognostic value of hypoxia PET in LASCCHN for LC. Outcome after hypoxia DE appears promising and may support the concept of DE. Slow accrual and premature closure may partly be due to a high complexity of the study setup which needs to be considered for future multicenter trials.
Collapse
Affiliation(s)
- Stefan Welz
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Matthias Reimold
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christian La Fougère
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Alber
- Section for Medical Physics, Department of Radiation Oncology, Heidelberg University, Heidelberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University of Munich, Germany; Department of Radiation Oncology, LMU Munich, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
17
|
PET/CT. PET Clin 2022; 17:297-305. [DOI: 10.1016/j.cpet.2021.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
18
|
The value of plasma hypoxia markers for predicting imaging-based hypoxia in patients with head-and-neck cancers undergoing definitive chemoradiation. Clin Transl Radiat Oncol 2022; 33:120-127. [PMID: 35243023 PMCID: PMC8881198 DOI: 10.1016/j.ctro.2022.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Higher osteopontin plasma levels correlate with more hypoxic tumors at baseline. Increased baseline osteopontin levels are associated with residual tumor hypoxia. Absent early hypoxia response is linked with higher VEGF and CTGF levels in week 5. Plasma hypoxic markers may serve as biomarkers favoring radiotherapy personalization.
Background Methods Results Conclusion
Collapse
|
19
|
Evensen ME, Furre T, Malinen E, Løndalen AM, Dale E. Mucosa-sparing dose painting of head and neck cancer. Acta Oncol 2022; 61:141-145. [PMID: 34991431 DOI: 10.1080/0284186x.2021.2022200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
| | - Torbjørn Furre
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | | | - Einar Dale
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
20
|
Fu S, Li Y, Han Y, Wang H, Chen Y, Yan O, He Q, Ma H, Liu L, Liu F. Diffusion-weighted MRI-guided dose painting in patients with locoregionally advanced nasopharyngeal carcinoma treated with induction chemotherapy plus concurrent chemoradiotherapy: a randomized, controlled clinical trial. Int J Radiat Oncol Biol Phys 2022; 113:101-113. [PMID: 35074433 DOI: 10.1016/j.ijrobp.2021.12.175] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 12/23/2021] [Accepted: 12/28/2021] [Indexed: 12/24/2022]
Abstract
PURPOSES We hypothesized that diffusion-weighted MRI (DWI)-guided dose-painting intensity modulated radiation therapy (DP-IMRT) is associated with improved local tumor control and survival in locoregionally advanced nasopharyngeal carcinoma (NPC). The purpose of this randomized study was to compare the efficacy and toxicity of DWI-guided DP-IMRT to conventional MRI-based IMRT in locoregional advanced NPC. METHODS A total of 260 NPC patients with stage III-IVa disease were randomly assigned in a 1:1 ratio to receive induction chemotherapy followed by chemoradiotherapy by DWI-guided DP-IMRT (group A, n = 130) or conventional MRI-based IMRT (group B, n = 130) in this prospective clinical trial. In group A, subvolume GTVnx-DWI (gross tumor volume of nasopharynx in DWI) was defined as the areas within the GTVnx (gross tumor volume of nasopharynx) with an apparent diffusion coefficient (ADC) below the mean ADC (ADC < mean) according to MRI before induction chemotherapy. The dose to GTVnx-DWI was escalated to 75.2 Gy/32 Fx in patients with T1-2 disease and to 77.55 Gy/33 Fx in those with T3-4 disease in 2.35 Gy per fraction. In group B, PGTVnx was irradiated at 70.4-72.6 Gy/32-33 Fx in 2.2 Gy per fraction. This trial is registered with chictr.org.cn (number). RESULTS A total of 260 patients were included in the trial (130 patients in group A and 130 in group B). Complete response rates after chemoradiotherapy were 99.2% (129/130) and 93.8% (122/130) in groups A and B, respectively (P=0.042). At a median follow-up of 25 months, DWI-guided DP-IMRT was associated with improved 2-year disease-free survival (DFS, 93.6% [95% CI, 88.1% to 99.1%] vs. 87.5% [95% CI, 81.4% to 93.6%], P = 0.015), local recurrence-free survival (LRFS, 100% [95% CI, not applicable (NA)] vs. 91.3% [95% CI, 85.4% to 97.2%]), locoregional recurrence-free survival (LRRFS, 95.8% [95% CI, NA] vs. 91.3% [95% CI, 85.4% to 97.2%]), distant metastasis-free survival (DMFS, 97.8% [95% CI, NA] vs. 90.9% [95% CI, 85.8% to 96.0%]), and overall survival (OS, 100% [95% CI, NA] vs. 94.5% [95% CI, 89.2% to 99.8%]). There were 0 and 3 patients had local-only recurrences in group A and B, respectively. The most common site of first failure in each arm was distant organ failure. No statistically significant differences in acute and late toxic effects were observed. Multivariate analyses showed that dose painting (DWI-guided DP-IMRT vs conventional MRI-based IMRT without DP) was associated with DFS, LRFS, LRRFS and DMFS. EBV DNA level was associated with DFS and LRRFS. CONCLUSIONS DWI-guided DP-IMRT plus chemotherapy is associated with a disease-free survival benefit compared with conventional MRI-based IMRT among patients with locoregionally advanced NPC without increasing acute toxicity.
Collapse
Affiliation(s)
- Shengnan Fu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanxian Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yaqian Han
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hui Wang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Key Laboratory of Translational Radiation Oncology, Hunan Province, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanzhu Chen
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ouying Yan
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qian He
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Hongzhi Ma
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lin Liu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Feng Liu
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
21
|
Clasen K, Gani C, Schroeder C, Riess O, Zips D, Schöffski O, Clasen S. The patients view on genetics and functional imaging for precision medicine: a willingness-to-pay analysis. Per Med 2022; 19:103-112. [PMID: 34984920 DOI: 10.2217/pme-2021-0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Purpose: Willingness-to-pay (WTP) analyses can support allocation processes considering the patients preferences in personalized medicine. However, genetic testing especially might imply ethical concerns that have to be considered. Methods: A WTP questionnaire was designed to compare preferences for imaging and genetic testing in cancer patients and to evaluate potential ethical concerns. Results: Comparing the options of imaging and genetics showed comparable WTP values. Ethical concerns about genetic testing seemed to be minor. Treatment success was the top priority irrespective of the diagnostic modality. In general, the majority of patients considered personalized medicine to be beneficial. Conclusion: Most patients valued personalized approaches and rated the benefits of precision medicine of overriding importance irrespective of modality or ethical concerns.
Collapse
Affiliation(s)
- Kerstin Clasen
- Department of Radiation Oncology, Medical Faculty & University Hospital, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, Tübingen, 72076, Germany
| | - Cihan Gani
- Department of Radiation Oncology, Medical Faculty & University Hospital, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, Tübingen, 72076, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) partner site Tübingen, Hoppe-Seyler-Straße 3, Tübingen, 72076, Germany
| | - Christopher Schroeder
- Institute of Medical Genetics & Applied Genomics, Medical Faculty & University Hospital, Eberhard Karls University Tübingen, Calwerstraße 7, Tübingen, 72076, Germany
| | - Olaf Riess
- Institute of Medical Genetics & Applied Genomics, Medical Faculty & University Hospital, Eberhard Karls University Tübingen, Calwerstraße 7, Tübingen, 72076, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Medical Faculty & University Hospital, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 3, Tübingen, 72076, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ) partner site Tübingen, Hoppe-Seyler-Straße 3, Tübingen, 72076, Germany
| | - Oliver Schöffski
- Department of Health Management, Friedrich-Alexander-University Erlangen-Nuremberg (FAU), Lange Gasse 20, Nuremberg, 90403, Germany
| | - Stephan Clasen
- Department of Diagnostic & Interventional Radiology, District Hospital Reutlingen, Steinenbergstraße 31, Reutlingen, 72764, Germany
| |
Collapse
|
22
|
den bossche VV, Zaryouh H, Vara-Messler M, Vignau J, Machiels JP, Wouters A, Schmitz S, Corbet C. Microenvironment-driven intratumoral heterogeneity in head and neck cancers: clinical challenges and opportunities for precision medicine. Drug Resist Updat 2022; 60:100806. [DOI: 10.1016/j.drup.2022.100806] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
|
23
|
Wegge M, Dok R, Nuyts S. Hypoxia and Its Influence on Radiotherapy Response of HPV-Positive and HPV-Negative Head and Neck Cancer. Cancers (Basel) 2021; 13:5959. [PMID: 34885069 PMCID: PMC8656584 DOI: 10.3390/cancers13235959] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cancers are a heterogeneous group of cancers that arise from the upper aerodigestive tract. Etiologically, these tumors are linked to alcohol/tobacco abuse and infections with high-risk human papillomavirus (HPV). HPV-positive HNSCCs are characterized by a different biology and also demonstrate better therapy response and survival compared to alcohol/tobacco-related HNSCCs. Despite this advantageous therapy response and the clear biological differences, all locally advanced HNSCCs are treated with the same chemo-radiotherapy schedules. Although we have a better understanding of the biology of both groups of HNSCC, the biological factors associated with the increased radiotherapy response are still unclear. Hypoxia, i.e., low oxygen levels because of an imbalance between oxygen demand and supply, is an important biological factor associated with radiotherapy response and has been linked with HPV infections. In this review, we discuss the effects of hypoxia on radiotherapy response, on the tumor biology, and the tumor microenvironment of HPV-positive and HPV-negative HNSCCs by pointing out the differences between these two tumor types. In addition, we provide an overview of the current strategies to detect and target hypoxia.
Collapse
Affiliation(s)
- Marilyn Wegge
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
| | - Rüveyda Dok
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
| | - Sandra Nuyts
- Laboratory of Experimental Radiotherapy, Department of Oncology, University of Leuven, 3000 Leuven, Belgium; (M.W.); (R.D.)
- Department of Radiation Oncology, Leuven Cancer Institute, UZ Leuven, 3000 Leuven, Belgium
| |
Collapse
|
24
|
Rühle A, Wiedenmann N, Fennell JT, Mix M, Ruf J, Stoian R, Thomsen AR, Vaupel P, Baltas D, Grosu AL, Nicolay NH. Interleukin-6 as surrogate marker for imaging-based hypoxia dynamics in patients with head-and-neck cancers undergoing definitive chemoradiation-results from a prospective pilot trial. Eur J Nucl Med Mol Imaging 2021; 49:1650-1660. [PMID: 34773163 PMCID: PMC8940848 DOI: 10.1007/s00259-021-05602-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
Purpose Intratumoral hypoxia increases resistance of head-and-neck squamous cell carcinoma (HNSCC) to radiotherapy. [18F]FMISO PET imaging enables noninvasive hypoxia monitoring, though requiring complex logistical efforts. We investigated the role of plasma interleukin-6 (IL-6) as potential surrogate parameter for intratumoral hypoxia in HNSCC using [18F]FMISO PET/CT as reference. Methods Within a prospective trial, serial blood samples of 27 HNSCC patients undergoing definitive chemoradiation were collected to analyze plasma IL-6 levels. Intratumoral hypoxia was assessed in treatment weeks 0, 2, and 5 using [18F]FMISO PET/CT imaging. The association between PET-based hypoxia and IL-6 was examined using Pearson’s correlation and multiple regression analyses, and the diagnostic power of IL-6 for tumor hypoxia response prediction was determined with receiver-operating characteristic analyses. Results Mean IL-6 concentrations were 15.1, 19.6, and 31.0 pg/mL at baseline, week 2 and week 5, respectively. Smoking (p=0.050) and reduced performance status (p=0.011) resulted in higher IL-6 levels, whereas tumor (p=0.427) and nodal stages (p=0.334), tumor localization (p=0.439), and HPV status (p=0.294) had no influence. IL-6 levels strongly correlated with the intratumoral hypoxic subvolume during treatment (baseline: r=0.775, p<0.001; week 2: r=0.553, p=0.007; week 5: r=0.734, p<0.001). IL-6 levels in week 2 were higher in patients with absent early tumor hypoxia response (p=0.016) and predicted early hypoxia response (AUC=0.822, p=0.031). Increased IL-6 levels at week 5 resulted in a trend towards reduced progression-free survival (p=0.078) and overall survival (p=0.013). Conclusion Plasma IL-6 is a promising surrogate marker for tumor hypoxia dynamics in HNSCC patients and may facilitate hypoxia-directed personalized radiotherapy concepts. Trial registration The prospective trial was registered in the German Clinical Trial Register (DRKS00003830). Registered 20 August 2015 Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05602-x.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jamina T Fennell
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Raluca Stoian
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas R Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
25
|
Wright P, Arnesen MR, Lønne PI, Suilamo S, Silvoniemi A, Dale E, Minn H, Malinen E. Repeatability of hypoxia dose painting by numbers based on EF5-PET in head and neck cancer. Acta Oncol 2021; 60:1386-1391. [PMID: 34184605 DOI: 10.1080/0284186x.2021.1944663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Hypoxia dose painting is a radiotherapy technique to increase the dose to hypoxic regions of the tumour. Still, the clinical effect relies on the reproducibility of the hypoxic region shown in the medical image. 18F-EF5 is a hypoxia tracer for positron emission tomography (PET), and this study investigated the repeatability of 18F-EF5-based dose painting by numbers (DPBN) in head and neck cancer (HNC). MATERIALS AND METHODS Eight HNC patients undergoing two 18F-EF5-PET/CT sessions (A and B) before radiotherapy were included. A linear conversion of PET signal intensity to radiotherapy dose prescription was employed and DPBN treatment plans were created using the image basis acquired at each PET/CT session. Also, plan A was recalculated on the image basis for session B. Voxel-by-voxel Pearson's correlation and quality factor were calculated to assess the DPBN plan quality and repeatability. RESULTS The mean (SD) correlation coefficient between DPBN prescription and plan was 0.92 (0.02) and 0.93 (0.02) for sessions A and B, respectively, with corresponding quality factors of 0.02 (0.002) and 0.02 (0.003), respectively. The mean correlation between dose prescriptions at day A and B was 0.72 (0.13), and 0.77 (0.12) for the corresponding plans. A mean correlation of 0.80 (0.08) was found between plan A, recalculated on image basis B, and plan B. CONCLUSION Hypoxia DPBN planning based on 18F-EF5-PET/CT showed high repeatability. This illustrates that 18F-EF5-PET provides a robust target for dose painting.
Collapse
Affiliation(s)
- Pauliina Wright
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | | | - Per-Ivar Lønne
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
| | - Sami Suilamo
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland
- Department of Medical Physics, Turku University Hospital, Turku, Finland
| | - Antti Silvoniemi
- Department of Otorhinolaryngology-Head and Neck Surgery, Turku University Hospital, Turku PET Centre, University of Turku, Turku, Finland
| | - Einar Dale
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Heikki Minn
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku PET Centre, University of Turku, Turku, Finland
| | - Eirik Malinen
- Department of Medical Physics, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Strahlenther Onkol 2021; 197:1-23. [PMID: 34259912 DOI: 10.1007/s00066-021-01812-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Christoph Henkenberens
- Department of Radiotherapy and Special Oncology, Medical School Hannover, Hannover, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany.
| |
Collapse
|
27
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Nuklearmedizin 2021; 60:326-343. [PMID: 34261141 DOI: 10.1055/a-1525-7029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | | | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiotherapy and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | | |
Collapse
|
28
|
Carles M, Fechter T, Grosu AL, Sörensen A, Thomann B, Stoian RG, Wiedenmann N, Rühle A, Zamboglou C, Ruf J, Martí-Bonmatí L, Baltas D, Mix M, Nicolay NH. 18F-FMISO-PET Hypoxia Monitoring for Head-and-Neck Cancer Patients: Radiomics Analyses Predict the Outcome of Chemo-Radiotherapy. Cancers (Basel) 2021; 13:3449. [PMID: 34298663 PMCID: PMC8303992 DOI: 10.3390/cancers13143449] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/06/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia is associated with radiation resistance and can be longitudinally monitored by 18F-fluoromisonidazole (18F-FMISO)-PET/CT. Our study aimed at evaluating radiomics dynamics of 18F-FMISO-hypoxia imaging during chemo-radiotherapy (CRT) as predictors for treatment outcome in head-and-neck squamous cell carcinoma (HNSCC) patients. We prospectively recruited 35 HNSCC patients undergoing definitive CRT and longitudinal 18F-FMISO-PET/CT scans at weeks 0, 2 and 5 (W0/W2/W5). Patients were classified based on peritherapeutic variations of the hypoxic sub-volume (HSV) size (increasing/stable/decreasing) and location (geographically-static/geographically-dynamic) by a new objective classification parameter (CP) accounting for spatial overlap. Additionally, 130 radiomic features (RF) were extracted from HSV at W0, and their variations during CRT were quantified by relative deviations (∆RF). Prediction of treatment outcome was considered statistically relevant after being corrected for multiple testing and confirmed for the two 18F-FMISO-PET/CT time-points and for a validation cohort. HSV decreased in 64% of patients at W2 and in 80% at W5. CP distinguished earlier disease progression (geographically-dynamic) from later disease progression (geographically-static) in both time-points and cohorts. The texture feature low grey-level zone emphasis predicted local recurrence with AUCW2 = 0.82 and AUCW5 = 0.81 in initial cohort (N = 25) and AUCW2 = 0.79 and AUCW5 = 0.80 in validation cohort. Radiomics analysis of 18F-FMISO-derived hypoxia dynamics was able to predict outcome of HNSCC patients after CRT.
Collapse
Affiliation(s)
- Montserrat Carles
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Tobias Fechter
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Anca L. Grosu
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Arnd Sörensen
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Benedikt Thomann
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Raluca G. Stoian
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nicole Wiedenmann
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Alexander Rühle
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Constantinos Zamboglou
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Juri Ruf
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Luis Martí-Bonmatí
- La Fe Health Research Institute, Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), 46026 Valencia, Spain;
| | - Dimos Baltas
- Department of Radiation Oncology, Division of Medical Physics, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; (T.F.); (B.T.); (D.B.)
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
| | - Michael Mix
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Nuclear Medicine, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nils H. Nicolay
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Partner Site Freiburg of the German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (A.L.G.); (A.S.); (R.G.S.); (N.W.); (A.R.); (C.Z.); (J.R.); (M.M.); (N.H.N.)
- Department of Radiation Oncology, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
29
|
Ferini G, Valenti V, Tripoli A, Illari SI, Molino L, Parisi S, Cacciola A, Lillo S, Giuffrida D, Pergolizzi S. Lattice or Oxygen-Guided Radiotherapy: What If They Converge? Possible Future Directions in the Era of Immunotherapy. Cancers (Basel) 2021; 13:cancers13133290. [PMID: 34209192 PMCID: PMC8268715 DOI: 10.3390/cancers13133290] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/31/2022] Open
Abstract
Palliative radiotherapy has a great role in the treatment of large tumor masses. However, treating a bulky disease could be difficult, especially in critical anatomical areas. In daily clinical practice, short course hypofractionated radiotherapy is delivered in order to control the symptomatic disease. Radiation fields generally encompass the entire tumor mass, which is homogeneously irradiated. Recent technological advances enable delivering a higher radiation dose in small areas within a large mass. This goal, previously achieved thanks to the GRID approach, is now achievable using the newest concept of LATTICE radiotherapy (LT-RT). This kind of treatment allows exploiting various radiation effects, such as bystander and abscopal effects. These events may be enhanced by the concomitant use of immunotherapy, with the latter being ever more successfully delivered in cancer patients. Moreover, a critical issue in the treatment of large masses is the inhomogeneous intratumoral distribution of well-oxygenated and hypo-oxygenated areas. It is well known that hypoxic areas are more resistant to the killing effect of radiation, hence the need to target them with higher aggressive doses. This concept introduces the "oxygen-guided radiation therapy" (OGRT), which means looking for suitable hypoxic markers to implement in PET/CT and Magnetic Resonance Imaging. Future treatment strategies are likely to involve combinations of LT-RT, OGRT, and immunotherapy. In this paper, we review the radiobiological rationale behind a potential benefit of LT-RT and OGRT, and we summarize the results reported in the few clinical trials published so far regarding these issues. Lastly, we suggest what future perspectives may emerge by combining immunotherapy with LT-RT/OGRT.
Collapse
Affiliation(s)
- Gianluca Ferini
- REM Radioterapia, Viagrande, I-95029 Catania, Italy; (V.V.); (A.T.)
- Correspondence: ; Tel.: +39-095-789-4581
| | - Vito Valenti
- REM Radioterapia, Viagrande, I-95029 Catania, Italy; (V.V.); (A.T.)
| | | | | | - Laura Molino
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali Università di Messina, I-98100 Messina, Italy; (L.M.); (S.P.); (A.C.); (S.L.); (S.P.)
| | - Silvana Parisi
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali Università di Messina, I-98100 Messina, Italy; (L.M.); (S.P.); (A.C.); (S.L.); (S.P.)
| | - Alberto Cacciola
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali Università di Messina, I-98100 Messina, Italy; (L.M.); (S.P.); (A.C.); (S.L.); (S.P.)
| | - Sara Lillo
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali Università di Messina, I-98100 Messina, Italy; (L.M.); (S.P.); (A.C.); (S.L.); (S.P.)
| | - Dario Giuffrida
- Medical Oncology Unit, Mediterranean Institute of Oncology, Viagrande, I-95029 Catania, Italy;
| | - Stefano Pergolizzi
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali Università di Messina, I-98100 Messina, Italy; (L.M.); (S.P.); (A.C.); (S.L.); (S.P.)
| |
Collapse
|
30
|
Telarovic I, Wenger RH, Pruschy M. Interfering with Tumor Hypoxia for Radiotherapy Optimization. J Exp Clin Cancer Res 2021; 40:197. [PMID: 34154610 PMCID: PMC8215813 DOI: 10.1186/s13046-021-02000-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/30/2021] [Indexed: 12/11/2022] Open
Abstract
Hypoxia in solid tumors is an important predictor of treatment resistance and poor clinical outcome. The significance of hypoxia in the development of resistance to radiotherapy has been recognized for decades and the search for hypoxia-targeting, radiosensitizing agents continues. This review summarizes the main hypoxia-related processes relevant for radiotherapy on the subcellular, cellular and tissue level and discusses the significance of hypoxia in radiation oncology, especially with regard to the current shift towards hypofractionated treatment regimens. Furthermore, we discuss the strategies to interfere with hypoxia for radiotherapy optimization, and we highlight novel insights into the molecular pathways involved in hypoxia that might be utilized to increase the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Irma Telarovic
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Martin Pruschy
- Laboratory for Applied Radiobiology, Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
| |
Collapse
|
31
|
Wang YF, Tadimalla S, Hayden AJ, Holloway L, Haworth A. Artificial intelligence and imaging biomarkers for prostate radiation therapy during and after treatment. J Med Imaging Radiat Oncol 2021; 65:612-626. [PMID: 34060219 DOI: 10.1111/1754-9485.13242] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/18/2021] [Accepted: 05/02/2021] [Indexed: 12/15/2022]
Abstract
Magnetic resonance imaging (MRI) is increasingly used in the management of prostate cancer (PCa). Quantitative MRI (qMRI) parameters, derived from multi-parametric MRI, provide indirect measures of tumour characteristics such as cellularity, angiogenesis and hypoxia. Using Artificial Intelligence (AI), relevant information and patterns can be efficiently identified in these complex data to develop quantitative imaging biomarkers (QIBs) of tumour function and biology. Such QIBs have already demonstrated potential in the diagnosis and staging of PCa. In this review, we explore the role of these QIBs in monitoring treatment response during and after PCa radiotherapy (RT). Recurrence of PCa after RT is not uncommon, and early detection prior to development of metastases provides an opportunity for salvage treatments with curative intent. However, the current method of monitoring treatment response using prostate-specific antigen levels lacks specificity. QIBs, derived from qMRI and developed using AI techniques, can be used to monitor biological changes post-RT providing the potential for accurate and early diagnosis of recurrent disease.
Collapse
Affiliation(s)
- Yu-Feng Wang
- Institute of Medical Physics, School of Physics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
| | - Sirisha Tadimalla
- Institute of Medical Physics, School of Physics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Amy J Hayden
- Sydney West Radiation Oncology, Westmead Hospital, Wentworthville, New South Wales, Australia
- Faculty of Medicine, Western Sydney University, Sydney, New South Wales, Australia
- Faculty of Medicine, Health & Human Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Lois Holloway
- Institute of Medical Physics, School of Physics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, Australia
- Liverpool and Macarthur Cancer Therapy Centre, Liverpool Hospital, Liverpool, New South Wales, Australia
| | - Annette Haworth
- Institute of Medical Physics, School of Physics, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
32
|
Bogowicz M, Pavic M, Riesterer O, Finazzi T, Garcia Schüler H, Holz-Sapra E, Rudofsky L, Basler L, Spaniol M, Ambrusch A, Hüllner M, Guckenberger M, Tanadini-Lang S. Targeting Treatment Resistance in Head and Neck Squamous Cell Carcinoma - Proof of Concept for CT Radiomics-Based Identification of Resistant Sub-Volumes. Front Oncol 2021; 11:664304. [PMID: 34123824 PMCID: PMC8191457 DOI: 10.3389/fonc.2021.664304] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/06/2021] [Indexed: 12/18/2022] Open
Abstract
Purpose Radiomics has already been proposed as a prognostic biomarker in head and neck cancer (HNSCC). However, its predictive power in radiotherapy has not yet been studied. Here, we investigated a local radiomics approach to distinguish between tumor sub-volumes with different levels of radiosensitivity as a possible target for radiation dose intensification. Materials and Methods Of 40 patients (n=28 training and n=12 validation) with biopsy confirmed locally recurrent HNSCC, pretreatment contrast-enhanced CT images were registered with follow-up PET/CT imaging allowing identification of controlled (GTVcontrol) vs non-controlled (GTVrec) tumor sub-volumes on pretreatment imaging. A bi-regional model was built using radiomic features extracted from pretreatment CT in the GTVrec and GTVcontrol to differentiate between those regions. Additionally, concept of local radiomics was implemented to perform detection task. The original tumor volume was divided into sub-volumes with no prior information on the location of recurrence. Radiomic features from those sub-volumes were then used to detect recurrent sub-volumes using multivariable logistic regression. Results Radiomic features extracted from non-controlled regions differed significantly from those in controlled regions (training AUC = 0.79 CI 95% 0.66 - 0.91 and validation AUC = 0.88 CI 95% 0.72 – 1.00). Local radiomics analysis allowed efficient detection of non-controlled sub-volumes both in the training AUC = 0.66 (CI 95% 0.56 – 0.75) and validation cohort 0.70 (CI 95% 0.53 – 0.86), however performance of this model was inferior to bi-regional model. Both models indicated that sub-volumes characterized by higher heterogeneity were linked to tumor recurrence. Conclusion Local radiomics is able to detect sub-volumes with decreased radiosensitivity, associated with location of tumor recurrence in HNSCC in the pre-treatment CT imaging. This proof of concept study, indicates that local CT radiomics can be used as predictive biomarker in radiotherapy and potential target for dose intensification.
Collapse
Affiliation(s)
- Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Oliver Riesterer
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Centre for Radiation Oncology KSA-KSB, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Tobias Finazzi
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Helena Garcia Schüler
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Edna Holz-Sapra
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Leonie Rudofsky
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lucas Basler
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Manon Spaniol
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andreas Ambrusch
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stephanie Tanadini-Lang
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Kashihara T, Nakamura S, Murakami N, Ito K, Matsumoto Y, Kobayashi K, Omura G, Mori T, Honma Y, Kubo Y, Okamoto H, Takahashi K, Inaba K, Okuma K, Igaki H, Nakayama Y, Kato K, Matsumoto F, Yoshimoto S, Itami J. Initial Experience of Intentional Internal High-Dose Policy Volumetric Modulated Arc Therapy of Neck Lymph Node Metastases ≥ 2 cm in Patients With Head and Neck Squamous Cell Carcinoma. Front Oncol 2021; 11:651409. [PMID: 33987086 PMCID: PMC8112241 DOI: 10.3389/fonc.2021.651409] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Background and Purpose Most locoregional recurrences after definitive radiotherapy for head and neck squamous cell carcinoma (HNSCC) develop "in-field." Dose escalation while sparing organs at risk can be a good solution for improving local control without increasing adverse effects. This study investigated the safety and effectiveness of volumetric modulated arc therapy (VMAT) using intentionally internal high-dose policy (IIHDP) to treat neck lymph node metastases (NLNM) ≥ 2 cm in HNSCC patients. Materials and Methods We analyzed 71 NLNM from 51 HNSCC patients who had received definitive radiotherapy to treat NLNM ≥ 2 cm using the VMAT technique in our institution between February 2017 and August 2019. Thirty-seven NLNM from 25 patients were treated using IIHDP VMAT (group A), and 34 NLNM from 27 patients were treated with homogeneous-dose distribution policy (HDDP) VMAT (group B). One patient with three NLNM had one lymph node assigned to group A and the other two to group B. Adverse events and local recurrence-free survival (LRFS) was compared between the two groups. Results In the median follow-up period of 527 days, there were no significant differences between the groups in terms of dermatitis or mucositis ≥ grade 2/3, but LRFS was significantly longer in group A (p = 0.007). In the Cox regression analysis after adjustment for the propensity score, group A also showed an apparently superior LFRS. Conclusion Our initial experience of IIHDP VMAT suggested that IIHDP VMAT to treat HNSCC neck lymph node metastases measuring ≥ 2 cm was feasible and possibly led to better local control than HDDP VMAT.
Collapse
Affiliation(s)
- Tairo Kashihara
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Satoshi Nakamura
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Murakami
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Kimiteru Ito
- Department of Radiology in National Cancer Center Hospital, Tokyo, Japan
| | - Yoshifumi Matsumoto
- Department of Head and Neck Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Kenya Kobayashi
- Department of Head and Neck Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Go Omura
- Department of Head and Neck Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Taisuke Mori
- Department of Pathology and Clinical Laboratories in National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Honma
- Department of Head and Neck Medical Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Kubo
- Department of Radiology in National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Okamoto
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Kana Takahashi
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Koji Inaba
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Kae Okuma
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Hiroshi Igaki
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Nakayama
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck Medical Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Fumihiko Matsumoto
- Department of Head and Neck Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Oncology in National Cancer Center Hospital, Tokyo, Japan
| | - Jun Itami
- Department of Radiation Therapy in National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
34
|
Khan R, Seltzer M. PET Imaging of Tumor Hypoxia in Head and Neck Cancer: A Primer for Neuroradiologists. Neuroimaging Clin N Am 2021; 30:325-339. [PMID: 32600634 DOI: 10.1016/j.nic.2020.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Tumor hypoxia is a known independent prognostic factor for adverse patient outcomes in those with head and neck cancer. Areas of tumor hypoxia have been found to be more radiation resistant than areas of tumor with normal oxygenation levels. Hypoxia imaging may serve to help identify the best initial treatment option and to assess intratreatment monitoring of tumor response in case treatment changes can be made. PET imaging is the gold standard method for imaging tumor hypoxia, with 18F-fluoromisonidazole the most extensively studied hypoxic imaging tracer. Newer tracers also show promise.
Collapse
Affiliation(s)
- Rihan Khan
- Department of Radiology, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA.
| | - Marc Seltzer
- Department of Radiology, Dartmouth-Hitchcock Medical Center, 1 Medical Center Drive, Lebanon, NH 03756, USA
| |
Collapse
|
35
|
Flaus A, Nevesny S, Guy JB, Sotton S, Magné N, Prévot N. Positron emission tomography for radiotherapy planning in head and neck cancer: What impact? Nucl Med Commun 2021; 42:234-243. [PMID: 33252513 DOI: 10.1097/mnm.0000000000001329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PET-computed tomography (CT) plays a growing role to guide target volume delineation for head and neck cancer in radiation oncology. Pretherapeutic [18F]FDG PET-CT adds information to morphological imaging. First, as a whole-body imaging modality, it reveals regional or distant metastases that induce major therapeutic changes in more than 10% of the cases. Moreover, it allows better pathological lymph node selection which improves overall regional control and overall survival. Second, locally, it allows us to define the metabolic tumoral volume, which is a reliable prognostic feature for survival outcome. [18F]FDG PET-CT-based gross tumor volume (GTV) is on average significantly smaller than GTV based on CT. Nevertheless, the overlap is incomplete and more evaluation of composite GTV based on PET and GTV based on CT are needed. However, in clinical practice, the study showed that using GTV PET alone for treatment planning was similar to using GTVCT for local control and dose distribution was better as a dose to organs at risk significantly decreased. In addition to FDG, pretherapeutic PET could give access to different biological tumoral volumes - thanks to different tracers - guiding heterogeneous dose delivery (dose painting concept) to resistant subvolumes. During radiotherapy treatment, follow-up [18F]FDG PET-CT revealed an earlier and more important diminution of GTV than other imaging modality. It may be a valuable support for adaptative radiotherapy as a new treatment plan with a significant impact on dose distribution became possible. Finally, additional studies are required to prospectively validate long-term outcomes and lower toxicity resulting from the use of PET-CT in treatment planning.
Collapse
Affiliation(s)
- Anthime Flaus
- Service de Médecine Nucléaire, Centre Hospitalier Universitaire de Saint-Etienne, St Etienne
| | - Stéphane Nevesny
- Département de Radiothérapie, Institut de Cancérologie de la Loire-Lucien Neuwirth, St Priest en Jarez
| | - Jean-Baptiste Guy
- Département de Radiothérapie, Institut de Cancérologie de la Loire-Lucien Neuwirth, St Priest en Jarez
- UMR CNRS 5822/IN2P3, IPNL, PRISME, Laboratoire de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Université Lyon 1, Oullins Cedex
| | - Sandrine Sotton
- Department of Research and Teaching, Lucien Neuwirth Cancer Institute, Saint-Priest-en-Jarez, University Departement of Research and Teaching
| | - Nicolas Magné
- Département de Radiothérapie, Institut de Cancérologie de la Loire-Lucien Neuwirth, St Priest en Jarez
- UMR CNRS 5822/IN2P3, IPNL, PRISME, Laboratoire de Radiobiologie Cellulaire et Moléculaire, Faculté de Médecine Lyon-Sud, Université Lyon 1, Oullins Cedex
| | - Nathalie Prévot
- Service de Médecine Nucléaire, Centre Hospitalier Universitaire de Saint-Etienne, St Etienne
- INSERM U 1059 Sainbiose, Université Jean Monnet, Saint-Etienne, France
| |
Collapse
|
36
|
Fever range whole body hyperthermia for re-irradiation of head and neck squamous cell carcinomas: Final results of a prospective study. Oral Oncol 2021; 116:105240. [PMID: 33626457 DOI: 10.1016/j.oraloncology.2021.105240] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/22/2021] [Accepted: 02/13/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Fever-range whole body hyperthermia (FRWBH) has been shown to improve tumor oxygenation in vivo. A prospective pilot study addressed the question if addition of FRWBH to re-irradiation is feasible in recurrent head and neck squamous cell carcinomas (HNSCC) with unfavorable prognostic features. MATERIALS AND METHODS The study completed accrual with the recruitment of ten patients between April 2018 and March 2020. Re-irradiation was administered using volumetric arc hyperfractionated radiotherapy with bi-daily 1.2 Gray (Gy) single fractions and a total dose of 66 Gy to all macroscopic tumor lesions. Concomitant chemotherapy consisted mostly of cisplatin (7 patients). FRWBH was scheduled weekly during re-irradiation. The study was registered in the clinicaltrials.gov database (NCT03547388). RESULTS Only five patients received all cycles of FRWBH. Poor patient compliance, active infections during treatment and study restrictions due to the Covid-19 pandemic were the main reasons for omitting FRWBH. No increase of acute toxicity was observed by FRWBH. Exploratory evaluation of outcome data suggests that FRWBH treatment according to protocol does not seem to have a detrimental effect on tumor control or survival and might even increase treatment efficacy. CONCLUSION FRWBH is difficult to apply concomitant to re-irradiation in HNSCC. No excess toxicity was observed in patients receiving FRWBH and exploratory analyses suggest potential anti-tumor activity and decreased patient-reported depression scores after FRWBH.
Collapse
|
37
|
Lu J, Zhang C, Yang X, Yao XJ, Zhang Q, Sun XC. Synthesis and Preliminary Evaluation of a Novel 18F-Labeled 2-Nitroimidazole Derivative for Hypoxia Imaging. Front Oncol 2021; 10:572097. [PMID: 33604284 PMCID: PMC7884749 DOI: 10.3389/fonc.2020.572097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/09/2020] [Indexed: 11/13/2022] Open
Abstract
Objective Hypoxia is prevalent in tumors and plays a pivotal role in resistance to chemoradiotherapy. 18F-MISO (18F-labeled fluoromisonidazole) is currently the preferred choice of PET hypoxia tracers in clinical practice, but has severe disadvantages involving complex labeling methods and low efficient imaging due to lipophilicity. We aimed to design a novel nitroimidazole derivative labeled by 18F via a chelation technique to detect hypoxic regions and provide a basis for planning radiotherapy. Materials and Methods First, we synthesized a 2-nitroimidazole precursor, 2-[4-(carboxymethyl)-7-[2-(2-(2-nitro-1H-imidazol-1-yl)acetamido)ethyl]-1,4,7-triazanonan-1-yl]acetic acid (NOTA-NI). For 18F-labeling, a 18F solution was reacted with a mixture of AlCl3 and NOTA-NI at pH 3.5 and 100°C for 20 min, and the radiochemical purity and stability were evaluated. Biological behaviors of Al18F-NOTA-NI were analyzed by an uptake study in ECA109 normoxic and hypoxic cells, and a biodistribution study and microPET imaging in ECA109 xenografted mice. Results Al18F-NOTA-NI required a straightforward and efficient labeling procedure compared with 18F-MISO. The uptake values were distinctly higher in hypoxic tumor cells. Animal studies revealed that the imaging agent was principally excreted via the kidneys. Due to hydrophilicity, the radioactivities in blood and muscle were decreased, and we could clearly distinguish xenografted tumors from para-carcinoma tissue by PET imaging. Conclusions The nitroimidazole tracer Al18F-NOTA-NI steadily accumulated in hypoxic areas in tumors and was rapidly eliminated from normal tissue. It appears to be a promising candidate for hypoxia imaging with high sensitivity and resolution.
Collapse
Affiliation(s)
- Jing Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chi Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi-Juan Yao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qun Zhang
- Department of Health Promotion Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin-Chen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
38
|
Tumor Hypoxia as a Barrier in Cancer Therapy: Why Levels Matter. Cancers (Basel) 2021; 13:cancers13030499. [PMID: 33525508 PMCID: PMC7866096 DOI: 10.3390/cancers13030499] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/22/2021] [Accepted: 01/23/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Hypoxia is a common feature of solid tumors and associated with poor outcome in most cancer types and treatment modalities, including radiotherapy, chemotherapy, surgery and, most likely, immunotherapy. Emerging strategies, such as proton therapy and combination therapies with radiation and hypoxia targeted drugs, provide new opportunities to overcome the hypoxia barrier and improve therapeutic outcome. Hypoxia is heterogeneously distributed both between and within tumors and shows large variations across patients not only in prevalence, but importantly, also in level. To best exploit the emerging strategies, a better understanding of how individual hypoxia levels from mild to severe affect tumor biology is vital. Here, we discuss our current knowledge on this topic and how we should proceed to gain more insight into the field. Abstract Hypoxia arises in tumor regions with insufficient oxygen supply and is a major barrier in cancer treatment. The distribution of hypoxia levels is highly heterogeneous, ranging from mild, almost non-hypoxic, to severe and anoxic levels. The individual hypoxia levels induce a variety of biological responses that impair the treatment effect. A stronger focus on hypoxia levels rather than the absence or presence of hypoxia in our investigations will help development of improved strategies to treat patients with hypoxic tumors. Current knowledge on how hypoxia levels are sensed by cancer cells and mediate cellular responses that promote treatment resistance is comprehensive. Recently, it has become evident that hypoxia also has an important, more unexplored role in the interaction between cancer cells, stroma and immune cells, influencing the composition and structure of the tumor microenvironment. Establishment of how such processes depend on the hypoxia level requires more advanced tumor models and methodology. In this review, we describe promising model systems and tools for investigations of hypoxia levels in tumors. We further present current knowledge and emerging research on cellular responses to individual levels, and discuss their impact in novel therapeutic approaches to overcome the hypoxia barrier.
Collapse
|
39
|
Imaging Hypoxia. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00074-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
40
|
Rabasco Meneghetti A, Zwanenburg A, Leger S, Leger K, Troost EG, Linge A, Lohaus F, Schreiber A, Kalinauskaite G, Tinhofer I, Guberina N, Guberina M, Balermpas P, von der Grün J, Ganswindt U, Belka C, Peeken JC, Combs SE, Böke S, Zips D, Krause M, Baumann M, Löck S. Definition and validation of a radiomics signature for loco-regional tumour control in patients with locally advanced head and neck squamous cell carcinoma. Clin Transl Radiat Oncol 2021. [DOI: 10.1016/j.ctro.2020.11.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
41
|
Masson I, Dutreix M, Supiot S. [Innovation in radiotherapy in 2021]. Bull Cancer 2020; 108:42-49. [PMID: 33303195 DOI: 10.1016/j.bulcan.2020.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Ingrid Masson
- Département de radiothérapie, Institut de cancérologie de l'Ouest René-Gauducheau, boulevard Jacques-Monod, 44805 Saint-Herblain, France
| | - Marie Dutreix
- Institut Curie, Université PSL, CNRS, Inserm, UMR 3347; Université Paris Sud, Université Paris-Saclay, 91405 Orsay, France
| | - Stéphane Supiot
- Département de radiothérapie, Institut de cancérologie de l'Ouest René-Gauducheau, boulevard Jacques-Monod, 44805 Saint-Herblain, France; Centre de Recherche en Cancéro-Immunologie Nantes/Angers (CRCINA, UMR 892 Inserm), Institut de Recherche en Santé de l'Université de Nantes, Nantes cedex 1, France.
| |
Collapse
|
42
|
Rogasch J, Beck M, Stromberger C, Hofheinz F, Ghadjar P, Wust P, Budach V, Amthauer H, Tinhofer I, Furth C, Walter-Rittel TC, Zschaeck S. PET measured hypoxia and MRI parameters in re-irradiated head and neck squamous cell carcinomas: findings of a prospective pilot study. F1000Res 2020; 9:1350. [PMID: 33796277 PMCID: PMC7970429 DOI: 10.12688/f1000research.27303.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 01/04/2023] Open
Abstract
Background: Tumor hypoxia measured by dedicated tracers like [
18F]fluoromisonidazole (FMISO) is a well-established prognostic factor in head and neck squamous cell carcinomas (HNSCC) treated with definitive chemoradiation (CRT). However, prevalence and characteristics of positron emission tomography (PET) measured hypoxia in patients with relapse after previous irradiation is missing. Here we report imaging findings of a prospective pilot study in HNSCC patients treated with re-irradiation. Methods: In 8 patients with recurrent HNSCC, diagnosed at a median of 18 months after initial radiotherapy/CRT, [
18F]fluorodeoxyglucose (FDG)-PET/CT (n=8) and FMISO-PET/MRI (n=7) or FMISO-PET/CT (n=1) were performed. Static FMISO-PET was performed after 180 min. MRI sequences in PET/MRI included diffusion-weighted imaging with apparent diffusion coefficient (ADC) values and contrast enhanced T1w imaging (StarVIBE). Lesions (primary tumor recurrence, 4; cervical lymph node, 1; both, 3) were delineated on FDG-PET and FMISO-PET data using a background-adapted threshold-based method. SUV
max and SUV
mean in FDG- and FMISO-PET were derived, as well as maximum tumor-to-muscle ratio (TMR
max) and hypoxic volume with 1.6-fold muscle SUV
mean (HV
1.6) in FMISO-PET. Intensity of lesional contrast enhancement was rated relative to contralateral normal tissue. Average ADC values were derived from a 2D region of interest in the tumor. Results: In FMISO-PET, median TMR
max was 1.7 (range: 1.1-1.8). Median HV
1.6 was 0.05 ml (range: 0-7.3 ml). Only in 2/8 patients, HV
1.6 was ≥1.0 ml. In FDG-PET, median SUV
max was 9.3 (range: 5.0-20.1). On contrast enhanced imaging four lesions showed decreased and four lesions increased contrast enhancement compared to non-pathologic reference tissue. Median average ADC was 1,060 ×10
6 mm
2/s (range: 840-1,400 ×10
6 mm
2/s). Conclusions: This pilot study implies that hypoxia detectable by FMISO-PET may not be as prevalent as expected among loco-regional recurrent, HPV negative HNSCC. ADC values were only mildly reduced, and contrast enhancement was variable. The results require confirmation in larger sample sizes.
Collapse
Affiliation(s)
- Julian Rogasch
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Stromberger
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Pirus Ghadjar
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Wust
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ingeborg Tinhofer
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thula C Walter-Rittel
- Department of Diagnostic and Interventional Radiology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
43
|
Pigorsch SU, Kampfer S, Oechsner M, Mayinger MC, Mozes P, Devecka M, Kessel KK, Combs SE, Wilkens JJ. Report on planning comparison of VMAT, IMRT and helical tomotherapy for the ESCALOX-trial pre-study. Radiat Oncol 2020; 15:253. [PMID: 33138837 PMCID: PMC7607845 DOI: 10.1186/s13014-020-01693-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background The ESCALOX trial was designed as a multicenter, randomized prospective dose escalation study for head and neck cancer. Therefore, feasibility of treatment planning via different treatment planning systems (TPS) and radiotherapy (RT) techniques is essential. We hypothesized the comparability of dose distributions for simultaneous integrated boost (SIB) volumes respecting the constraints by different TPS and RT techniques. Methods CT data sets of the first six patients (all male, mean age: 61.3 years) of the pre-study (up to 77 Gy) were used for comparison of IMRT, VMAT, and helical tomotherapy (HT). Oropharynx was the primary tumor location. Normalization of the three step SIB (77 Gy, 70 Gy, 56 Gy) was D95% = 77 Gy. Coverage (CVF), healthy tissue conformity index (HTCI), conformation number (CN), and dose homogeneity (HI) were compared for PTVs and conformation index (COIN) for parotids. Results All RT techniques achieved good coverage. For SIB77Gy, CVF was best for IMRT and VMAT, HT achieved highest CN followed by VMAT and IMRT. HT reached good HTCI value, and HI compared to both other techniques. For SIB70Gy, CVF was best by IMRT. HTCI favored HT, consequently CN as well. HI was slightly better for HT. For SIB56Gy, CVF resulted comparably. Conformity favors VMAT as seen by HTCI and CN. Dmean of ipsilateral and contralateral parotids favor HT. Conclusion Different TPS for dose escalation reliably achieved high plan quality. Despite the very good results of HT planning for coverage, conformity, and homogeneity, the TPS also achieved acceptable results for IMRT and VMAT. Trial registration ClinicalTrials.gov Identifier: NCT 01212354, EudraCT-No.: 2010-021139-15. ARO: ARO 14-01
Collapse
Affiliation(s)
- Steffi U Pigorsch
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany.
| | - Severin Kampfer
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| | - Markus Oechsner
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| | - Michael C Mayinger
- Department of Radiation Oncology, University Hospital Zurich, Rämistrasse 100, Zurich, Switzerland
| | - Petra Mozes
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| | - Michal Devecka
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| | - Kerstin K Kessel
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| | - Stephanie E Combs
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany.,Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Ingolstädter Landstraße 1, Neuherberg, Germany
| | - Jan J Wilkens
- Department of Radiation Oncology, Technical University of Munich (TUM), School of Medicine, Klinikum Rechts Der Isar, Ismaninger Straße 22, 81675, Munich, Germany
| |
Collapse
|
44
|
Starke S, 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, Troost EGC, Krause M, Baumann M, Löck S. 2D and 3D convolutional neural networks for outcome modelling of locally advanced head and neck squamous cell carcinoma. Sci Rep 2020; 10:15625. [PMID: 32973220 PMCID: PMC7518264 DOI: 10.1038/s41598-020-70542-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/20/2020] [Indexed: 12/14/2022] Open
Abstract
For treatment individualisation of patients with locally advanced head and neck squamous cell carcinoma (HNSCC) treated with primary radiochemotherapy, we explored the capabilities of different deep learning approaches for predicting loco-regional tumour control (LRC) from treatment-planning computed tomography images. Based on multicentre cohorts for exploration (206 patients) and independent validation (85 patients), multiple deep learning strategies including training of 3D- and 2D-convolutional neural networks (CNN) from scratch, transfer learning and extraction of deep autoencoder features were assessed and compared to a clinical model. Analyses were based on Cox proportional hazards regression and model performances were assessed by the concordance index (C-index) and the model's ability to stratify patients based on predicted hazards of LRC. Among all models, an ensemble of 3D-CNNs achieved the best performance (C-index 0.31) with a significant association to LRC on the independent validation cohort. It performed better than the clinical model including the tumour volume (C-index 0.39). Significant differences in LRC were observed between patient groups at low or high risk of tumour recurrence as predicted by the model ([Formula: see text]). This 3D-CNN ensemble will be further evaluated in a currently ongoing prospective validation study once follow-up is complete.
Collapse
Affiliation(s)
- Sebastian Starke
- Helmholtz-Zentrum Dresden - Rossendorf, Department of Information Services and Computing, Dresden, Germany.
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany.
| | - 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
| | - 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas Schreiber
- Department of Radiotherapy, Hospital Dresden-Friedrichstadt, Dresden, Germany
| | - Goda Kalinauskaite
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Berlin, Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Inge Tinhofer
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Berlin, Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Nika Guberina
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Essen, Essen, Germany
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Maja Guberina
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Essen, Essen, Germany
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Panagiotis Balermpas
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Frankfurt, Frankfurt, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Jens von der Grün
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Frankfurt, Frankfurt, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ute Ganswindt
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Munich, Germany
- Department of Radiation Oncology, Ludwig-Maximilians-Universität, Munich, Germany
- Clinical Cooperation Group, Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
- Department of Radiation Oncology, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Claus Belka
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Munich, Germany
- Department of Radiation Oncology, Ludwig-Maximilians-Universität, Munich, Germany
- Clinical Cooperation Group, Personalized Radiotherapy in Head and Neck Cancer, Helmholtz Zentrum, Munich, Germany
| | - Jan C Peeken
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Munich, Germany
- Department of Radiation Oncology, Technische Universität München, Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Stephanie E Combs
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Munich, Germany
- Department of Radiation Oncology, Technische Universität München, Munich, Germany
- Institute of Radiation Medicine (IRM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Simon Boeke
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Daniel Zips
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Tübingen, Tübingen, Germany
- Department of Radiation Oncology, Faculty of Medicine and University Hospital Tübingen, Eberhard Karls Universität Tübingen, 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Esther G C Troost
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, 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, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Steffen Löck
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
45
|
Positron Emission Tomography and Molecular Imaging of Head and Neck Malignancies. CURRENT RADIOLOGY REPORTS 2020. [DOI: 10.1007/s40134-020-00366-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
MRI- and CT-determined changes of dysphagia / aspiration-related structures (DARS) during and after radiotherapy. PLoS One 2020; 15:e0237501. [PMID: 32877418 PMCID: PMC7467287 DOI: 10.1371/journal.pone.0237501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 07/09/2020] [Indexed: 11/22/2022] Open
Abstract
Purpose The concept of dysphagia/aspiration-related structures (DARS) was developed against the background of severe late side effects of radiotherapy (RT) for head and neck cancer (HNC). DARS can be delineated on CT scans, but with a better morphological discrimination on magnetic resonance imaging (MRI). Swallowing function was analyzed by use of patient charts and prospective investigations and questionnaires. Method Seventeen HNC patients treated with intensity-modulated radiotherapy (IMRT) ± chemotherapy between 5/2012 – 8/2015 were included. Planning CT (computed tomography) scans and MRIs (magnetic resonance imaging) prior, during 40 Gray (Gy) radiotherapy and posttreatment were available and co-registered to delineate DARS. The RT dose of each DARS was calculated. Five patients were investigated posttreatment for swallowing function and assessed by means of various questionnaires for quality of life (QoL), swallowing, and voice function. Results By retrospective comparison of DARS volume, a significant change in four of eight DARS was detected over time. Three increased and one diminished. The risk of posttreatment dysphagia rose by every 1Gy above the mean dose (D mean) of RT to DARS. 7.5 was the risk factor for dysphagia in the first 6 months, reducing to 4.7 for months 6-12 posttreatment. For all five patients of the prospective part of swallowing investigations, a function disturbance was detected. These results were in contrast to the self-assessment of patients by questionnaires. There was neither a dose dependency of D mean DARS volume changes over time nor of dysphonia and no correlation between volume changes, dysphagia or dysphonia. Conclusion Delineation of DARS on MRI co-registered to planning CT gave the opportunity to differentiate morphology better than by CT alone. Due to the small number of patients with complete MRI scans over time, we failed to detect a dose dependency of DARS and swallowing and voice disorder posttreatment.
Collapse
|
47
|
Nicolay NH, Rühle A, Wiedenmann N, Niedermann G, Mix M, Weber WA, Baltas D, Werner M, Kayser G, Grosu AL. Lymphocyte Infiltration Determines the Hypoxia-Dependent Response to Definitive Chemoradiation in Head-and-Neck Cancer: Results from a Prospective Imaging Trial. J Nucl Med 2020; 62:471-478. [PMID: 32859699 DOI: 10.2967/jnumed.120.248633] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia in head-and-neck squamous cell carcinoma (HNSCC) leads to an immunosuppressive microenvironment and reduces the response to radiotherapy. In this prospective imaging trial, we investigated potential interactions between functional hypoxia imaging and infiltrating lymphocyte levels as a potential predictor for treatment response in HNSCC patients. Methods: In total, 49 patients receiving definitive chemoradiation for locally advanced HNSCCs underwent pretherapeutic biopsies and peritherapeutic hypoxia imaging using 18F-misonidazole PET at weeks 0, 2, and 5 during chemoradiation. Hematoxylin-eosin and immunohistochemical stainings for tumor-infiltrating lymphocytes, tissue-based hypoxia, and microvascular markers were analyzed and correlated with the longitudinal hypoxia dynamics and patient outcomes. Results: High levels of tumor-infiltrating total lymphocytes correlated with superior locoregional control (LRC) (hazard ratio [HR], 0.279; P = 0.011) and progression-free survival (PFS) (HR, 0.276; P = 0.006). Similarly, early resolution of 18F-misonidazole PET-detected tumor hypoxia quantified by 18F-misonidazole dynamics between weeks 0 and 2 of chemoradiation was associated with improved LRC (HR, 0.321; P = 0.015) and PFS (HR, 0.402; P = 0.043). Outcomes in the favorable early hypoxia resolution subgroup significantly depended on infiltrating lymphocyte counts, with patients who showed both an early hypoxia response and high lymphocyte infiltration levels exhibiting significantly improved LRC (HR, 0.259; P = 0.036) and PFS (HR, 0.242; P = 0.017) compared with patients with an early hypoxia response but low lymphocyte counts. These patients exhibited oncologic results comparable to those of patients with no hypoxia response within the first 2 wk of chemoradiation. Conclusion: This analysis established a clinical hypoxia-immune score that predicted treatment responses and outcomes in HNSCC patients undergoing chemoradiation and may help to devise novel concepts for biology-driven personalization of chemoradiation.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany .,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany; and
| | - Dimos Baltas
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Martin Werner
- German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Gian Kayser
- German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
48
|
Saksø M, Mortensen LS, Primdahl H, Johansen J, Kallehauge J, Hansen CR, Overgaard J. Influence of FAZA PET hypoxia and HPV-status for the outcome of head and neck squamous cell carcinoma (HNSCC) treated with radiotherapy: Long-term results from the DAHANCA 24 trial (NCT01017224). Radiother Oncol 2020; 151:126-133. [PMID: 32805273 DOI: 10.1016/j.radonc.2020.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/30/2020] [Accepted: 08/11/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Hypoxic tumor volumes can be visualized with 18F-FAZA PET/CT. In head and neck squamous cell carcinoma (HNSCC), hypoxia is important for the clinical outcome after primary radiotherapy (RT). The outcome is furthermore heavily influenced by the HPV/p16-positivity of oropharyngeal tumors (OPCp16+ tumors). The study purposes were (1) to report on locoregional failures within five years after primary RT in a prospective cohort stratified by both HPV/p16-status and PET hypoxia and (2) to characterize the failure site and the spatial association to PET hypoxia. MATERIAL AND METHODS From 2009 to 2011, 38 patients with non-metastatic SCC of the larynx, oro-, hypo- and nasopharynx completing primary RT were included in the prospective DAHANCA 24 trial (NCT01017224). Fifteen patients had OPCp16+ tumors. All were imaged with a static FAZA PET/CT prior to treatment. The hypoxia threshold was determined by a tumor-to-muscle ratio (TMR) of 1.6. Recurrences were documented histologically. Imaging of the recurrence was deformable fused with the pre-treatment FAZA PET/CT. The spatial information of recurrence- and hypoxic volumes were compared visually. RESULTS Sixteen patients had more hypoxic tumors (high tracer uptake, TMR ≥1.6) before treatment (42%). With a median follow-up of 7.8 years, nine locoregional recurrences were observed, of which seven were in patients with high-uptake tumors (44% and 9%, respectively, HR 5.8 [1.2-28.2]). The risk of locoregional recurrence was highest among patients with more hypoxic, non-OPCp16+ tumors (57% [21-94%]), with a risk difference of 45% [4-86%], when comparing to less hypoxic, non-OPCp16+ tumors. Eight patients had sufficient imaging of the recurrence for co-registration with the FAZA PET/CT. Six had hypoxic primary tumors, and in two, the recurrence was overlapping the baseline hypoxic subvolume. CONCLUSION HNSCC demonstrating a TMR ≥1.6 at baseline is significantly associated with treatment failure after primary RT. In addition to HPV/p16-status, FAZA PET/CT has potential for the selection of tumors requiring treatment intensification.
Collapse
Affiliation(s)
- Mette Saksø
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark.
| | | | - Hanne Primdahl
- Department of Oncology, Aarhus University Hospital, Denmark
| | | | | | | | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Denmark
| |
Collapse
|
49
|
Rühle A, Grosu AL, Wiedenmann N, Mix M, Stoian R, Niedermann G, Baltas D, Werner M, Weber WA, Kayser G, Nicolay NH. Hypoxia dynamics on FMISO-PET in combination with PD-1/PD-L1 expression has an impact on the clinical outcome of patients with Head-and-neck Squamous Cell Carcinoma undergoing Chemoradiation. Am J Cancer Res 2020; 10:9395-9406. [PMID: 32802199 PMCID: PMC7415814 DOI: 10.7150/thno.48392] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated hypoxia influences the radiation response of head-and-neck cancer (HNSCC) patients, and a lack of early hypoxia resolution during treatment considerably deteriorates outcomes. As the detrimental effects of hypoxia are partly related to the induction of an immunosuppressive microenvironment, we investigated the interaction between tumor hypoxia dynamics and the PD-1/PD-L1 axis in HNSCC patients undergoing chemoradiation and its relevance for patient outcomes in a prospective trial. Methods: 49 patients treated with definitive chemoradiation for locally advanced HNSCC were enrolled in this trial and received longitudinal hypoxia PET imaging using fluorine-18 misonidazole ([18F]FMISO) at weeks 0, 2 and 5 during treatment. Pre-therapeutic tumor biopsies were immunohistochemically analyzed regarding the PD-1/PD-L1 expression both on immune cells and on tumor cells, and potential correlations between the PD-1/PD-L1 axis and tumor hypoxia dynamics during chemoradiation were assessed using Spearman's rank correlations. Hypoxia dynamics during treatment were quantified by subtracting the standardized uptake value (SUV) index at baseline from the SUV values at weeks 2 or 5, whereby SUV index was defined as ratio of maximum tumor [18F]FMISO SUV to mean SUV in the contralateral sternocleidomastoid muscle (i.e. tumor-to-muscle ratio). The impact of the PD-1/PD-L1 expression alone and in combination with persistent tumor hypoxia on locoregional control (LRC), progression-free survival (PFS) and overall survival (OS) was examined using log-rank tests and Cox proportional hazards models. Results: Neither PD-L1 nor PD-1 expression levels on tumor-infiltrating immune cells influenced LRC (HR = 0.734; p = 0.480 for PD-L1, HR = 0.991; p = 0.989 for PD-1), PFS (HR = 0.813; p = 0.597 for PD-L1, HR = 0.796; p = 0.713 for PD-1) or OS (HR = 0.698; p = 0.405 for PD-L1, HR = 0.315; p = 0.265 for PD-1). However, patients with no hypoxia resolution between weeks 0 and 2 and PD-L1 expression on tumor cells, quantified by a tumor proportional score (TPS) of at least 1%, showed significantly worse LRC (HR = 3.374, p = 0.022) and a trend towards reduced PFS (HR = 2.752, p = 0.052). In the multivariate Cox regression analysis, the combination of absent tumor hypoxia resolution and high tumoral PD-L1 expression remained a significant prognosticator for impaired LRC (HR = 3.374, p = 0.022). On the other side, tumoral PD-L1 expression did not compromise the outcomes of patients whose tumor-associated hypoxia declined between week 0 and 2 during chemoradiation (LRC: HR = 1.186, p = 0.772, PFS: HR = 0.846, p = 0.766). Conclusion: In this exploratory analysis, we showed for the first time that patients with both persistent tumor-associated hypoxia during treatment and PD-L1 expression on tumor cells exhibited a worse outcome, while the tumor cells' PD-L1 expression did not influence the outcomes of patients with early tumor hypoxia resolution. While the results have to be validated in an independent cohort, these findings form a foundation to investigate the combination of hypoxic modification and immune checkpoint inhibitors for the unfavorable subgroup, moving forward towards personalized radiation oncology treatment.
Collapse
|
50
|
Her EJ, Haworth A, Reynolds HM, Sun Y, Kennedy A, Panettieri V, Bangert M, Williams S, Ebert MA. Voxel-level biological optimisation of prostate IMRT using patient-specific tumour location and clonogen density derived from mpMRI. Radiat Oncol 2020; 15:172. [PMID: 32660504 PMCID: PMC7805066 DOI: 10.1186/s13014-020-01568-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
AIMS This study aimed to develop a framework for optimising prostate intensity-modulated radiotherapy (IMRT) based on patient-specific tumour biology, derived from multiparametric MRI (mpMRI). The framework included a probabilistic treatment planning technique in the effort to yield dose distributions with an improved expected treatment outcome compared with uniform-dose planning approaches. METHODS IMRT plans were generated for five prostate cancer patients using two inverse planning methods: uniform-dose to the planning target volume and probabilistic biological optimisation for clinical target volume tumour control probability (TCP) maximisation. Patient-specific tumour location and clonogen density information were derived from mpMRI and geometric uncertainties were incorporated in the TCP calculation. Potential reduction in dose to sensitive structures was assessed by comparing dose metrics of uniform-dose plans with biologically-optimised plans of an equivalent level of expected tumour control. RESULTS The planning study demonstrated biological optimisation has the potential to reduce expected normal tissue toxicity without sacrificing local control by shaping the dose distribution to the spatial distribution of tumour characteristics. On average, biologically-optimised plans achieved 38.6% (p-value: < 0.01) and 51.2% (p-value: < 0.01) reduction in expected rectum and bladder equivalent uniform dose, respectively, when compared with uniform-dose planning. CONCLUSIONS It was concluded that varying the dose distribution within the prostate to take account for each patient's clonogen distribution was feasible. Lower doses to normal structures compared to uniform-dose plans was possible whilst providing robust plans against geometric uncertainties. Further validation in a larger cohort is warranted along with considerations for adaptive therapy and limiting urethral dose.
Collapse
Affiliation(s)
- E J Her
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, Australia.
| | - A Haworth
- Institute of Medical Physics, University of Sydney, Sydney, Australia
| | - H M Reynolds
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Y Sun
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - A Kennedy
- Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, Australia
| | - V Panettieri
- Alfred Health Radiation Oncology, Melbourne, Australia
| | - M Bangert
- Department of Medical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Physics in Radiation Oncology, Heidelberg Institute for Radiation Oncology, Heidelberg, Germany
| | - S Williams
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Division of Radiation Oncology and Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - M A Ebert
- School of Physics, Mathematics and Computing, University of Western Australia, Perth, Australia.,Department of Radiation Oncology, Sir Charles Gairdner Hospital, Perth, Australia.,5D Clinics, Perth, Australia
| |
Collapse
|