1
|
Soydal C, Demir B, Dursun E, Sutcu G, Celebioglu EC, Bilgic MS, Kucuk NO. Imaging of Hypoxia in Liver Tumors With 18 F-FMISO PET Before Selective Internal Radiotherapy With 90 Y Microspheres. Clin Nucl Med 2024; 49:e606-e607. [PMID: 38968590 DOI: 10.1097/rlu.0000000000005323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
ABSTRACT Hypoxia is a known cause of resistance to radiotherapy and chemotherapy. Although there are multiple studies in external radiation therapies based on hypoxia PET, the effect of hypoxia in radioembolization is largely unknown. Here we present 2 cases of hepatocellular carcinoma patients from a prospective study with different lesion characteristics on pretreatment 18 F-FMISO PET and varying responses on 18 F-FDG PET.
Collapse
|
2
|
Tárnoki ÁD, Tárnoki DL, Dąbrowska M, Knetki-Wróblewska M, Frille A, Stubbs H, Blyth KG, Juul AD. New developments in the imaging of lung cancer. Breathe (Sheff) 2024; 20:230176. [PMID: 38595936 PMCID: PMC11003524 DOI: 10.1183/20734735.0176-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/25/2024] [Indexed: 04/11/2024] Open
Abstract
Radiological and nuclear medicine methods play a fundamental role in the diagnosis and staging of patients with lung cancer. Imaging is essential in the detection, characterisation, staging and follow-up of lung cancer. Due to the increasing evidence, low-dose chest computed tomography (CT) screening for the early detection of lung cancer is being introduced to the clinical routine in several countries. Radiomics and radiogenomics are emerging fields reliant on artificial intelligence to improve diagnosis and personalised risk stratification. Ultrasound- and CT-guided interventions are minimally invasive methods for the diagnosis and treatment of pulmonary malignancies. In this review, we put more emphasis on the new developments in the imaging of lung cancer.
Collapse
Affiliation(s)
- Ádám Domonkos Tárnoki
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
- National Tumour Biology Laboratory, Oncologic Imaging and Invasive Diagnostic Centre, National Institute of Oncology, Budapest, Hungary
| | - Dávid László Tárnoki
- Medical Imaging Centre, Semmelweis University, Budapest, Hungary
- National Tumour Biology Laboratory, Oncologic Imaging and Invasive Diagnostic Centre, National Institute of Oncology, Budapest, Hungary
| | - Marta Dąbrowska
- Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Warsaw, Poland
| | | | - Armin Frille
- Department of Respiratory Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Harrison Stubbs
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Kevin G. Blyth
- Glasgow Pleural Disease Unit, Queen Elizabeth University Hospital, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
3
|
Hirata K, Watanabe S, Kitagawa Y, Kudo K. A Review of Hypoxia Imaging Using 18F-Fluoromisonidazole Positron Emission Tomography. Methods Mol Biol 2024; 2755:133-140. [PMID: 38319574 DOI: 10.1007/978-1-0716-3633-6_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Tumor hypoxia is an essential factor related to malignancy, prognosis, and resistance to treatment. Positron emission tomography (PET) is a modality that visualizes the distribution of radiopharmaceuticals administered into the body. PET imaging with [18F]fluoromisonidazole ([18F]FMISO) identifies hypoxic tissues. Unlike [18F]fluorodeoxyglucose ([18F]FDG)-PET, fasting is not necessary for [18F]FMISO-PET, but the waiting time from injection to image acquisition needs to be relatively long (e.g., 2-4 h). [18F]FMISO-PET images can be displayed on an ordinary commercial viewer on a personal computer (PC). While visual assessment is fundamental, various quantitative indices such as tumor-to-muscle ratio have also been proposed. Several novel hypoxia tracers have been invented to compensate for the limitations of [18F]FMISO.
Collapse
Affiliation(s)
- Kenji Hirata
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan.
- Department of Nuclear Medicine, Hokkaido University Hospital, Sapporo, Japan.
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| | - Shiro Watanabe
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Nuclear Medicine, Hokkaido University Hospital, Sapporo, Japan
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshimasa Kitagawa
- Oral Diagnosis and Medicine, Department of Oral Pathobiological Science, Graduate School of Dental Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kohsuke Kudo
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Department of Nuclear Medicine, Hokkaido University Hospital, Sapporo, Japan
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
4
|
Dos Santos SN, Wuest M, Jans HS, Woodfield J, Nario AP, Krys D, Dufour J, Glubrecht D, Bergman C, Bernardes ES, Wuest F. Comparison of three 18F-labeled 2-nitroimidazoles for imaging hypoxia in breast cancer xenografts: [ 18F]FBNA, [ 18F]FAZA and [ 18F]FMISO. Nucl Med Biol 2023; 124-125:108383. [PMID: 37651917 DOI: 10.1016/j.nucmedbio.2023.108383] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Tumour hypoxia is associated with increased metastasis, invasion, poor therapy response and prognosis. Most PET radiotracers developed and used for clinical hypoxia imaging belong to the 2-nitroimidazole family. Recently we have developed novel 2-nitroimidazole-derived PET radiotracer [18F]FBNA (N-(4-[18F]fluoro-benzyl)-2-(2-nitro-1H-imidazol-1-yl)-acet-amide), an 18F-labeled analogue of antiparasitic drug benznidazole. The present study aimed to analyze its radio-pharmacological properties and systematically compare its PET imaging profiles with [18F]FMISO and [18F]FAZA in preclinical triple-negative (MDA-MB231) and estrogen receptor-positive (MCF-7) breast cancer models. METHODS In vitro cellular uptake experiments were carried out in MDA-MB321 and MCF-7 cells under normoxic and hypoxic conditions. Metabolic stability in vivo was determined in BALB/c mice using radio-TLC analysis. Dynamic PET experiments over 3 h post-injection were performed in MDA-MB231 and MCF-7 tumour-bearing mice. Those PET data were used for kinetic modelling analysis utilizing the reversible two-tissue-compartment model. Autoradiography was carried out in tumour tissue slices and compared to HIF-1α immunohistochemistry. Detailed ex vivo biodistribution was accomplished in BALB/c mice, and this biodistribution data were used for dosimetry calculation. RESULTS Under hypoxic conditions in vitro cellular uptake was elevated in both cell lines, MCF-7 and MDA-MB231, for all three radiotracers. After intravenous injection, [18F]FBNA formed two radiometabolites, resulting in a final fraction of 65 ± 9 % intact [18F]FBNA after 60 min p.i. After 3 h p.i., [18F]FBNA tumour uptake reached SUV values of 0.78 ± 0.01 in MCF-7 and 0.61 ± 0.04 in MDA-MB231 tumours (both n = 3), representing tumour-to-muscle ratios of 2.19 ± 0.04 and 1.98 ± 0.15, respectively. [18F]FMISO resulted in higher tumour uptakes (SUV 1.36 ± 0.04 in MCF-7 and 1.23 ± 0.08 in MDA-MB231 (both n = 4; p < 0.05) than [18F]FAZA (0.66 ± 0.11 in MCF-7 and 0.63 ± 0.14 in MDA-MB231 (both n = 4; n.s.)), representing tumour-to-muscle ratios of 3.24 ± 0.30 and 3.32 ± 0.50 for [18F]FMISO, and 2.92 ± 0.74 and 3.00 ± 0.42 for [18F]FAZA, respectively. While the fraction per time of radiotracer entering the second compartment (k3) was similar within uncertainties for all three radiotracers in MDA-MB231 tumours, it was different in MCF-7 tumours. The ratios k3/(k3 + k2) and K1*k3/(k3 + k2) in MCF-7 tumours were also significantly different, indicating dissimilar fractions of radiotracer bound and trapped intracellularly: K1*k3/(k2 + k3) [18F]FMISO (0.0088 ± 0.001)/min, n = 4; p < 0.001) > [18F]FAZA (0.0052 ± 0.002)/min, n = 4; p < 0.01) > [18F]FBNA (0.003 ± 0.001)/min, n = 3). In contrast, in MDA-MB231 tumours, only K1 was significantly elevated for [18F]FMISO. However, this did not result in significant differences for K1*k3/(k2 + k3) for all three 2-nitroimidazoles in MDA-MB231 tumours. CONCLUSION Novel 2-nitroimidazole PET radiotracer [18F]FBNA showed uptake into hypoxic breast cancer cells and tumour tissue presumably associated with elevated HIF1-α expression. Systematic comparison of PET imaging performance with [18F]FMISO and [18F]FAZA in different types of preclinical breast cancer models revealed a similar tumour uptake profile for [18F]FBNA with [18F]FAZA and, despite its higher lipophilicity, still a slightly higher muscle tissue clearance compared to [18F]FMISO.
Collapse
Affiliation(s)
- Sofia Nascimento Dos Santos
- Radiopharmacy Center, Nuclear and Energy Research Institute (IPEN / CNEN - SP), CEP 05508-000 São Paulo, SP, Brazil
| | - Melinda Wuest
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Hans-Sonke Jans
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Jenilee Woodfield
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Arian Pérez Nario
- Radiopharmacy Center, Nuclear and Energy Research Institute (IPEN / CNEN - SP), CEP 05508-000 São Paulo, SP, Brazil
| | - Daniel Krys
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Jennifer Dufour
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Darryl Glubrecht
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Cody Bergman
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada
| | - Emerson Soares Bernardes
- Radiopharmacy Center, Nuclear and Energy Research Institute (IPEN / CNEN - SP), CEP 05508-000 São Paulo, SP, Brazil
| | - Frank Wuest
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton T6G 2R7, Alberta, Canada.
| |
Collapse
|
5
|
Djekidel M, Alsadi R, Abi Akl M, Bouhali O, O'Doherty J. Tumor microenvironment and fibroblast activation protein inhibitor (FAPI) PET: developments toward brain imaging. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1183471. [PMID: 39355017 PMCID: PMC11440979 DOI: 10.3389/fnume.2023.1183471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/03/2023] [Indexed: 10/03/2024]
Abstract
Fibroblast activation protein (FAP) is a type-II membrane bound glycoprotein specifically expressed by activated fibroblasts almost exclusively in pathological conditions including arthritis, fibrosis and cancer. FAP is overexpressed in cancer-associated fibroblasts (CAFs) located in tumor stroma, and is known to be involved in a variety of tumor-promoting activities such as angiogenesis, proliferation, resistance to chemotherapy, extracellular matrix remodeling and immunosuppression. In most cancer types, higher FAP expression is associated with worse clinical outcomes, leading to the hypothesis that FAP activity is involved in cancer development, cancer cell migration, and cancer spread. Recently, various high selectivity FAP inhibitors (FAPIs) have been developed and subsequently used for positron emission tomography (PET) imaging of different pathologies. Considering the paucity of widely available and especially mainstream reliable radioligands in brain cancer PET imaging, and the poor survival rates of patients with certain types of brain cancer such as glioblastoma, FAPI-PET represents a major development in enabling the detection of small primary or metastatic lesions in the brain due to its biological characteristics and low background accumulation. In this work, we aim to summarize the potential avenues for use of FAPI-PET, from the basic biological processes to oncologic imaging and with a main focus on brain imaging.
Collapse
Affiliation(s)
- Mehdi Djekidel
- Department of Radiology/Nuclear Medicine, Northwell Health, New York, NY, United States
| | - Rahaf Alsadi
- Division of Arts and Science, Texas A&M University at Qatar, Doha, Qatar
| | - Maya Abi Akl
- Division of Arts and Science, Texas A&M University at Qatar, Doha, Qatar
- Department of Electronics and Information Systems, Medical Image and Signal Processing (MEDISIP), Ghent University, Ghent, Belgium
| | - Othmane Bouhali
- Division of Arts and Science, Texas A&M University at Qatar, Doha, Qatar
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Jim O'Doherty
- Siemens Medical Solutions, Malvern, PA, United States
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, Charleston, SC, United States
- Radiography and Diagnostic Imaging, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Inada M, Nishimura Y, Hanaoka K, Nakamatsu K, Doi H, Uehara T, Komanishi M, Ishii K, Kaida H, Hosono M. Visualization of tumor hypoxia and re-oxygenation after stereotactic body radiation therapy in early peripheral lung cancer: A prospective study. Radiother Oncol 2023; 180:109491. [PMID: 36706956 DOI: 10.1016/j.radonc.2023.109491] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE In this study, fluoromisonidazole positron emission tomography (F-MISO PET/CT) was used to evaluate tumor hypoxia and re-oxygenation in patients with lung tumors treated with stereotactic body radiation therapy (SBRT). MATERIALS AND METHODS Patients with T1-2 N0 lung cancer were included in this study. The prescribed dose was 48-52 Gy in four fractions. F-MISO PET/CT was performed twice, before SBRT and 1-3 days after the first irradiation. The maximum standardized uptake value (SUVmax) and tumor/muscle ratio (TMR) were evaluated as indicators of hypoxia. The threshold for hypoxia was defined as a TMR of 1.30 or more. RESULTS Between 2016 and 2021, 15 patients were included. Pre-treatment tumor hypoxia was observed in nine tumors (60 %). TMR in all six tumors without pre-treatment hypoxia rose after single high-dose irradiation. In contrast, TMR in six of nine tumors with pre-treatment hypoxia dropped after irradiation, suggesting re-oxygenation. Although no local recurrence was noted, regional and/or distant relapses were seen in four patients (27 %). Of these, three had tumors with abnormal F-MISO uptake. The remaining patient had a tumor without signs of hypoxia on pre-treatment PET/CT. The 2-year progression free survival of patients with tumors with and without pre-treatment hypoxia were 30 % and 63 %, respectively (p = 0.319). CONCLUSION Tumor hypoxia reduced after single high-dose irradiation. Tumor with F-MISO uptake seems to be an unfavorable prognostic factor in lung SBRT.
Collapse
Affiliation(s)
- Masahiro Inada
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan.
| | - Yasumasa Nishimura
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Kohei Hanaoka
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Kiyoshi Nakamatsu
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Hiroshi Doi
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Takuya Uehara
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Mikihito Komanishi
- Division of Positron Emission Tomography, Institute of Advanced Clinical Medicine, Kindai University, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Kazunari Ishii
- Departments of Radiology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Hayato Kaida
- Departments of Radiology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| | - Makoto Hosono
- Departments of Radiation Oncology, Kindai University Faculty of Medicine, 377-2, Onohigashi, Osakasayama-city, Osaka, Japan
| |
Collapse
|
7
|
Gouel P, Decazes P, Vera P, Gardin I, Thureau S, Bohn P. Advances in PET and MRI imaging of tumor hypoxia. Front Med (Lausanne) 2023; 10:1055062. [PMID: 36844199 PMCID: PMC9947663 DOI: 10.3389/fmed.2023.1055062] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Tumor hypoxia is a complex and evolving phenomenon both in time and space. Molecular imaging allows to approach these variations, but the tracers used have their own limitations. PET imaging has the disadvantage of low resolution and must take into account molecular biodistribution, but has the advantage of high targeting accuracy. The relationship between the signal in MRI imaging and oxygen is complex but hopefully it would lead to the detection of truly oxygen-depleted tissue. Different ways of imaging hypoxia are discussed in this review, with nuclear medicine tracers such as [18F]-FMISO, [18F]-FAZA, or [64Cu]-ATSM but also with MRI techniques such as perfusion imaging, diffusion MRI or oxygen-enhanced MRI. Hypoxia is a pejorative factor regarding aggressiveness, tumor dissemination and resistance to treatments. Therefore, having accurate tools is particularly important.
Collapse
Affiliation(s)
- Pierrick Gouel
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Decazes
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Vera
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Isabelle Gardin
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Sébastien Thureau
- QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,Département de Radiothérapie, Centre Henri Becquerel, Rouen, France
| | - Pierre Bohn
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,*Correspondence: Pierre Bohn,
| |
Collapse
|
8
|
Zhu Z, Gong G, Wang L, Su Y, Lu J, Yin Y. Three-dimensional arterial spin labeling-guided dose painting radiotherapy for non-enhancing low-grade gliomas. Jpn J Radiol 2023; 41:335-346. [PMID: 36342645 PMCID: PMC9974719 DOI: 10.1007/s11604-022-01357-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE To investigate the feasibility and dosimetric characteristics of dose painting for non-enhancing low-grade gliomas (NE-LGGs) guided by three-dimensional arterial spin labeling (3D-ASL). MATERIALS AND METHODS Eighteen patients with NE-LGGs were enrolled. 3D-ASL, T2 fluid-attenuated inversion recovery (T2 Flair) and contrast-enhanced T1-weighted magnetic resonance images were obtained. The gross tumor volume (GTV) was delineated on the T2 Flair. The hyper-perfusion region of the GTV (GTV-ASL) was determined by 3D-ASL, and the GTV-SUB was obtained by subtracting the GTV-ASL from the GTV. The clinical target volume (CTV) was created by iso-tropically expanding the GTV by 1 cm. The planning target volume (PTV), PTV-ASL were obtained by expanding the external margins of the CTV, GTV-ASL, respectively. PTV-SUB was generated by subtracting PTV-ASL from PTV. Three plans were generated for each patient: a conventional plan (plan 1) without dose escalation delivering 95-110% of 45-60 Gy in 1.8-2 Gy fractions to the PTV and two dose-painting plans (plan 2 and plan 3) with dose escalating by 10-20% (range, 50-72 Gy) to the PTV-ASL based on plan 1. The plan 3 was obtained from plan 2 without the maximum dose constraint. The dosimetric differences among the three plans were compared. RESULTS The volume ratio of the PTV-ASL to the PTV was (23.49 ± 11.94)% (Z = - 3.724, P = 0.000). Compared with plan 1, D2%, D98% and Dmean of PTV-ASL increased by 14.67%,16.17% and 14.31% in plan2 and 19.84%,15.52% and 14.27% in plan3, respectively (P < 0.05); the D2% of the PTV and PTV-SUB increased by 11.89% and 8.34% in plan 2, 15.89% and 8.49% in plan 3, respectively (P < 0.05). The PTV coverages were comparable among the three plans (P > 0.05). In plan 2 and plan 3, the conformity indexes decreased by 18.60% and 12.79%; while the homogeneity index increased by 1.43 and 2 times (P < 0.05). Compared with plan 1, the D0.1 cc of brain stem and Dmax of optic chiasma were slightly increased in plan 2 and plan 3, and the absolute doses met the dose constraint. The doses of the other organs at risk (OARs) were similar among the three plans (P > 0.05). CONCLUSION The dose delivered to hyper-perfusion volume derived from 3D-ASL can increased by 10-20% while respecting the constraints to the OARs for NE-LGGs, which provides a basis for future individualized and precise radiotherapy, especially if the contrast agent cannot be injected or when contrast enhancement is uncertain.
Collapse
Affiliation(s)
- Zihong Zhu
- grid.488387.8Department of Oncology, Affiliated Hospital of Southwest Medical University, No.25 Taiping Street, Jiangyang District, Luzhou, 646000 Sichuan China ,grid.440144.10000 0004 1803 8437Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117 Shandong China
| | - Guanzhong Gong
- grid.440144.10000 0004 1803 8437Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117 Shandong China
| | - Lizhen Wang
- grid.440144.10000 0004 1803 8437Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117 Shandong China
| | - Ya Su
- grid.440144.10000 0004 1803 8437Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117 Shandong China
| | - Jie Lu
- grid.440144.10000 0004 1803 8437Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117 Shandong China
| | - Yong Yin
- Department of Oncology, Affiliated Hospital of Southwest Medical University, No.25 Taiping Street, Jiangyang District, Luzhou, 646000, Sichuan, China. .,Department of Radiation Oncology Physics and Technology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440 Jiyan Road, Huaiyin District, Jinan, 250117, Shandong, China.
| |
Collapse
|
9
|
Thureau S, Mallet R, Gouel P, Modzelewski R, Vera P. [What dose escalation in the treatment of locally advanced non-small cell lung cancer?]. Cancer Radiother 2022; 26:890-893. [PMID: 36075830 DOI: 10.1016/j.canrad.2022.07.004] [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: 06/24/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 11/29/2022]
Abstract
Despite significant therapeutic advances in the treatment of locally advanced inoperable non-small cell lung cancer (NSCLC), notably through adjuvant immunotherapy, the rate of therapeutic failure remains high. The use of positron emission tomography with fluorodeoxyglucose (FDG-PET), respiratory motion and intensity modulated radiotherapy (IMRT) have led to therapeutic improvements with reduced toxicity and better local control. The optimal dose to be delivered remains unknown due to discordant results of studies for almost 20 years and the way to define the area to benefit from a dose increase (whole volume, subvolume defined by pre- or per-radiotherapy PET).
Collapse
Affiliation(s)
- S Thureau
- Département de radiothérapie et de physique médicale, centre Henri-Becquerel, Rouen, France; Unité QuantIF LITIS EA 4108, université de Rouen, Normandie, France; Département d'imagerie, centre Henri-Becquerel, Rouen, France.
| | - R Mallet
- Département de radiothérapie et de physique médicale, centre Henri-Becquerel, Rouen, France
| | - P Gouel
- Département d'imagerie, centre Henri-Becquerel, Rouen, France
| | - R Modzelewski
- Unité QuantIF LITIS EA 4108, université de Rouen, Normandie, France; Département d'imagerie, centre Henri-Becquerel, Rouen, France
| | - P Vera
- Unité QuantIF LITIS EA 4108, université de Rouen, Normandie, France; Département d'imagerie, centre Henri-Becquerel, Rouen, France
| |
Collapse
|
10
|
Krarup MMK, Fischer BM, Christensen TN. New PET Tracers: Current Knowledge and Perspectives in Lung Cancer. Semin Nucl Med 2022; 52:781-796. [PMID: 35752465 DOI: 10.1053/j.semnuclmed.2022.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/11/2022]
Abstract
PET/CT with the tracer 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG) has improved diagnostic imaging in cancer and is routinely used for diagnosing, staging and treatment planning in lung cancer patients. However, pitfalls of [18F]FDG-PET/CT limit the use in specific settings. Additionally, lung cancer is still the leading cause of cancer associated death and has high risk of recurrence after curative treatment. These circumstances have led to the continuous search for more sensitive and specific PET tracers to optimize lung cancer diagnosis, staging, treatment planning and evaluation. The objective of this review is to present and discuss current knowledge and perspectives of new PET tracers for use in lung cancer. A literature search was performed on PubMed and clinicaltrials.gov, limited to the past decade, excluding case reports, preclinical studies and studies on established tracers such as [18F]FDG and DOTATE. The most relevant papers from the search were evaluated. Several tracers have been developed targeting specific tumor characteristics and hallmarks of cancer. A small number of tracers have been studied extensively and evaluated head-to-head with [18F]FDG-PET/CT, whereas others need further investigation and validation in larger clinical trials. At this moment, none of the tracers can replace [18F]FDG-PET/CT. However, they might serve as supplementary imaging methods to provide more knowledge about biological tumor characteristics and visualize intra- and inter-tumoral heterogeneity.
Collapse
Affiliation(s)
- Marie M K Krarup
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Copehagen University Hospital, Copenhagen, Denmark.
| | - Barbara M Fischer
- Department of Clinical Medicine, Faculty of Health, Univeristy of Copenhagen (UCPH), Copenhagen, Denmark; School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Tine N Christensen
- Department of Clinical Physiology and Nuclear Medicine, Rigshospitalet Copehagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
11
|
Hapdey S, Dubray B, Chastan M, Thureau S, Gouel P, Edet-Sanson A, Becker S, Vera P, Bouyeure-Petit AC. Respiratory gated multistatic PET reconstructions to delineate radiotherapy target volume in patients with mobile lung tumors. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:171-178. [PMID: 31922369 DOI: 10.23736/s1824-4785.19.03183-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
BACKGROUND PET-CT with 18F-FDG or other radiopharmaceuticals is a recommended tool to help the delineation of lung cancers candidate to radiotherapy. The motion artifacts caused by respiratory movements are reduced by 4D acquisitions. We introduced an extended reconstruction algorithm (multiple reconstruct register and average [multi-RRA]) which requires much shorter acquisition times than standard 4D PET-CT. Our aim was to evaluate the interest on multi-RRA images as an alternative of 3D and 4D PET-CT for the delineation of lung lesion. METHODS PET acquisitions synchronized to the respiratory signal were obtained in 18 patients with mobile lung tumors. We compared the tumor volumes delineated on Multi-RRA images to 3D and 4D PET-CT, considering the 4D CT as a reference. The tumor volumes were delineated and compared with topologic similarity indexes (Dice, Jaccard and overlap). RESULTS Twenty tumors were delineated. The volumes delineated with multi-RRA and 4D PET were not significantly different (mean difference of 0.2±0.7 mL). Comparison by pairs (Tukey-Kramer test) showed that 3D-PET volumes were significantly smaller than 4D-PET and multi-RRA volumes (P<0.001). Topologic similarity indexes with 4D-PET were slightly statistically higher with multi-RRA than with 3D-PET (Dice and Jaccard) or 4D-CT (Dice, Jaccard and Overlap). CONCLUSIONS The tumor volumes delineated on multi-RRA are similar to the volumes obtained with 4D PET, with shorter acquisition time.
Collapse
Affiliation(s)
- Sebastien Hapdey
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France -
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France -
| | - Bernard Dubray
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
- Department of Radiotherapy, Henri Becquerel Cancer Center, Rouen, France
| | - Mathieu Chastan
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France
| | - Sebastien Thureau
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
- Department of Radiotherapy, Henri Becquerel Cancer Center, Rouen, France
| | - Pierrick Gouel
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France
| | - Agathe Edet-Sanson
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
| | - Stéphanie Becker
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
| | - Pierre Vera
- Department of Nuclear Medicine, Henri Becquerel Cancer Center, Rouen, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
| | | |
Collapse
|
12
|
Daniel Humberto Pozza, Ramon Bezerra Andrade de Mello. Treatment Sequencing Strategies in Lung Cancer. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:323-336. [PMID: 35599008 PMCID: PMC9127753 DOI: 10.3779/j.issn.1009-3419.2022.104.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND The advances in the lung cancer screening methods and therapeutics, together with awareness towards deleterious habits, such as smoking, is increasing the overall survival with better quality of life for the patients. However, lung cancer is still one of the most common and fatal neoplasm with a high incidence and consequently burden to public health worldwide. Thus, based on guidelines and recent phases II and III clinical trials studies, this manuscript summarizes the current treatment sequencing strategies in lung cancer. METHODS A comprehensive search of related articles was performed focused on phases II and III clinical trials studies. RESULTS The lung cancer management should take into consideration the tumor characteristics, histology, molecular pathology and be discussed in a multidisciplinary team. Lung cancer treatment options comprises surgery whenever possible, radiotherapy associate with/or chemotherapy and immunotherapy as monotherapy, or combined with chemotherapy and best palliative care. CONCLUSIONS The screening predictability in more patients, smoking reduction, early diagnosis, better disease understanding and individualized, more effective and tolerable therapeutics are related to an increasing in overall survival and quality of life. In the near future improvement of personalized therapy in precision medicine is expected, enhancing new predictive biomarkers, optimal doses and optimal treatment sequencing as well as anti-cancer vaccines development.
Collapse
Affiliation(s)
- Daniel Humberto Pozza
- Department of Biomedicine, Faculty of Medicine and i3s, University of Porto, 4200-319 Porto, Portugal,Daniel Humberto Pozza, E-mail:
| | - Ramon Bezerra Andrade de Mello
- Discipline of Medical Oncology, Post-graduation Program in Medicine, Nine of July University (UNINOVE), São Paulo, Brazil./Nine of July Hospital, São Paulo, Brazil
| |
Collapse
|
13
|
Vaz SC, Adam JA, Delgado Bolton RC, Vera P, van Elmpt W, Herrmann K, Hicks RJ, Lievens Y, Santos A, Schöder H, Dubray B, Visvikis D, Troost EGC, de Geus-Oei LF. Joint EANM/SNMMI/ESTRO practice recommendations for the use of 2-[ 18F]FDG PET/CT external beam radiation treatment planning in lung cancer V1.0. Eur J Nucl Med Mol Imaging 2022; 49:1386-1406. [PMID: 35022844 PMCID: PMC8921015 DOI: 10.1007/s00259-021-05624-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/15/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE 2-[18F]FDG PET/CT is of utmost importance for radiation treatment (RT) planning and response monitoring in lung cancer patients, in both non-small and small cell lung cancer (NSCLC and SCLC). This topic has been addressed in guidelines composed by experts within the field of radiation oncology. However, up to present, there is no procedural guideline on this subject, with involvement of the nuclear medicine societies. METHODS A literature review was performed, followed by a discussion between a multidisciplinary team of experts in the different fields involved in the RT planning of lung cancer, in order to guide clinical management. The project was led by experts of the two nuclear medicine societies (EANM and SNMMI) and radiation oncology (ESTRO). RESULTS AND CONCLUSION This guideline results from a joint and dynamic collaboration between the relevant disciplines for this topic. It provides a worldwide, state of the art, and multidisciplinary guide to 2-[18F]FDG PET/CT RT planning in NSCLC and SCLC. These practical recommendations describe applicable updates for existing clinical practices, highlight potential flaws, and provide solutions to overcome these as well. Finally, the recent developments considered for future application are also reviewed.
Collapse
Affiliation(s)
- Sofia C. Vaz
- Nuclear Medicine Radiopharmacology, Champalimaud Centre for the Unkown, Champalimaud Foundation, Lisbon, Portugal
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Judit A. Adam
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Roberto C. Delgado Bolton
- Department of Diagnostic Imaging (Radiology) and Nuclear Medicine, University Hospital San Pedro and Centre for Biomedical Research of La Rioja (CIBIR), Logroño (La Rioja), Spain
| | - Pierre Vera
- Henri Becquerel Cancer Center, QuantIF-LITIS EA 4108, Université de Rouen, Rouen, France
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW – School for Oncology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Rodney J. Hicks
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Yolande Lievens
- Radiation Oncology Department, Ghent University Hospital and Ghent University, Ghent, Belgium
| | - Andrea Santos
- Nuclear Medicine Department, CUF Descobertas Hospital, Lisbon, Portugal
| | - Heiko Schöder
- Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, USA
| | - Bernard Dubray
- Department of Radiotherapy and Medical Physics, Centre Henri Becquerel, Rouen, France
- QuantIF-LITIS EA4108, University of Rouen, Rouen, France
| | | | - 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
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association / Helmholtz-Zentrum Dresden – Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Mäurer M, Käsmann L, Fleischmann DF, Oertel M, Jazmati D, Medenwald D. PET/CT-based adaptive radiotherapy of locally advanced non-small cell lung cancer in multicenter yDEGRO ARO 2017-01 cohort study. Radiat Oncol 2022; 17:29. [PMID: 35139856 PMCID: PMC8827193 DOI: 10.1186/s13014-022-01997-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/25/2022] [Indexed: 12/25/2022] Open
Abstract
Background Stage III non-small cell lung cancer (NSCLC) represents a highly heterogeneous disease and treatment burden. Advances in imaging modality show promising results for radiotherapy planning. In this multicentric study, we evaluated the impact of PET/CT-based radiotherapy planning on the prognosis of patients with stage III NSCLC.
Method and patients A retrospective observational cohort study (ARO 2017-01/NCT03055715) was conducted by the young DEGRO trial group of the German Society for Radiation Oncology (DEGRO) with the primary objective to assess the effect of tumour volume change during chemoradiotherapy and the secondary objective to assess the effect of treatment planning on survival. Three hundred forty-seven patients with stage III NSCLC treated at 21 university centers between January 2010 and December 2013 were enrolled in this trial. Patients received primary curative chemoradiotherapy with an intended dose of 50 Gy (hypofractionated) or > 60 Gy (normofractionated). To assess the effect of radiotherapy planning modality on overall survival, we used multivariate frailty models. Models were adjusted for gross tumor volume at the initiation of therapy, age, sex, simultaneous chemotherapy, lung comorbidities, RT dose and tumor grade. By considering the random effect, we can account for heterogeneity in survival and considered covariates within the model in relation to the study side. Results Patients were predominantly male (n = 269, 78.4%) with mainly adenocarcinoma (56.4%) and an average of 67.2 years. Adaptation of radiotherapy with consecutive reduction of irradiation volume showed no significant disadvantage for patient survival (HR = 1.21, 95% CI 0.89–1.64). The use of PET/CT co-registration in radiation planning tended to result in better oncologic outcomes, although no significant association could be shown (HR = 0.8, 95% CI 0.56–1.16). Centers with a consistent planning strategy performed better than those without a preferred planning method (0.62, 95% CI 0.41–0.94). Conclusion A consistent planning strategy has positive effects on overall survival. The use of PET/CT-based adaptive radiotherapy planning shows a similar survival prospect with the prospective of lower treatment volumes. In future research, toxicities need to be analysed in order to assess such reasoning.
Collapse
|
15
|
Read GH, Bailleul J, Vlashi E, Kesarwala AH. Metabolic response to radiation therapy in cancer. Mol Carcinog 2022; 61:200-224. [PMID: 34961986 PMCID: PMC10187995 DOI: 10.1002/mc.23379] [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: 08/11/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 11/11/2022]
Abstract
Tumor metabolism has emerged as a hallmark of cancer and is involved in carcinogenesis and tumor growth. Reprogramming of tumor metabolism is necessary for cancer cells to sustain high proliferation rates and enhanced demands for nutrients. Recent studies suggest that metabolic plasticity in cancer cells can decrease the efficacy of anticancer therapies by enhancing antioxidant defenses and DNA repair mechanisms. Studying radiation-induced metabolic changes will lead to a better understanding of radiation response mechanisms as well as the identification of new therapeutic targets, but there are few robust studies characterizing the metabolic changes induced by radiation therapy in cancer. In this review, we will highlight studies that provide information on the metabolic changes induced by radiation and oxidative stress in cancer cells and the associated underlying mechanisms.
Collapse
Affiliation(s)
- Graham H. Read
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Justine Bailleul
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Erina Vlashi
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, California
| | - Aparna H. Kesarwala
- Department of Radiation Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
16
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [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: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
17
|
Kennedy WR, DeWees TA, Acharya S, Mahmood M, Knutson NC, Goddu SM, Kavanaugh JA, Mitchell TJ, Rich KM, Kim AH, Leuthardt EC, Dowling JL, Dunn GP, Chicoine MR, Perkins SM, Huang J, Tsien CI, Robinson CG, Abraham CD. Internal dose escalation associated with increased local control for melanoma brain metastases treated with stereotactic radiosurgery. J Neurosurg 2021; 135:855-861. [PMID: 33307528 DOI: 10.3171/2020.7.jns192210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 07/09/2020] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The internal high-dose volume varies widely for a given prescribed dose during stereotactic radiosurgery (SRS) to treat brain metastases (BMs). This may be altered during treatment planning, and the authors have previously shown that this improves local control (LC) for non-small cell lung cancer BMs without increasing toxicity. Here, they seek to identify potentially actionable dosimetric predictors of LC after SRS for melanoma BM. METHODS The records of patients with unresected melanoma BM treated with single-fraction Gamma Knife RS between 2006 and 2017 were reviewed. LC was assessed on a per-lesion basis, defined as stability or a decrease in lesion size. Outcome-oriented approaches were utilized to determine optimal dichotomization for dosimetric variables relative to LC. Univariable and multivariable Cox regression analysis was implemented to evaluate the impact of collected parameters on LC. RESULTS Two hundred eighty-seven melanoma BMs in 79 patients were identified. The median age was 56 years (range 31-86 years). The median follow-up was 7.6 months (range 0.5-81.6 months), and the median survival was 9.3 months (range 1.3-81.6 months). Lesions were optimally stratified by volume receiving at least 30 Gy (V30) greater than or equal to versus less than 25%. V30 was ≥ and < 25% in 147 and 140 lesions, respectively. For all patients, 1-year LC was 83% versus 66% for V30 ≥ and < 25%, respectively (p = 0.001). Stratifying by volume, lesions 2 cm or less (n = 215) had 1-year LC of 82% versus 70% (p = 0.013) for V30 ≥ and < 25%, respectively. Lesions > 2 to 3 cm (n = 32) had 1-year LC of 100% versus 43% (p = 0.214) for V30 ≥ and < 25%, respectively. V30 was still predictive of LC even after controlling for the use of immunotherapy and targeted therapy. Radionecrosis occurred in 2.8% of lesions and was not significantly associated with V30. CONCLUSIONS For a given prescription dose, an increased internal high-dose volume, as indicated by measures such as V30 ≥ 25%, is associated with improved LC but not increased toxicity in single-fraction SRS for melanoma BM. Internal dose escalation is an independent predictor of improved LC even in patients receiving immunotherapy and/or targeted therapy. This represents a dosimetric parameter that is actionable at the time of treatment planning and warrants further evaluation.
Collapse
Affiliation(s)
| | - Todd A DeWees
- 2Department of Biomedical Statistics and Informatics, Mayo Clinic, Scottsdale, Arizona; and
| | - Sahaja Acharya
- 3Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | | | | | | | | | - Keith M Rich
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Albert H Kim
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Eric C Leuthardt
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Joshua L Dowling
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Gavin P Dunn
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | - Michael R Chicoine
- 4Neurosurgery, Washington University School of Medicine, St. Louis, Missouri
| | | | | | | | | | | |
Collapse
|
18
|
Thureau S, Modzelewski R, Bohn P, Hapdey S, Gouel P, Dubray B, Vera P. Comparison of Hypermetabolic and Hypoxic Volumes Delineated on [ 18F]FDG and [ 18F]Fluoromisonidazole PET/CT in Non-small-cell Lung Cancer Patients. Mol Imaging Biol 2021; 22:764-771. [PMID: 31432388 DOI: 10.1007/s11307-019-01422-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
PURPOSE The high rates of failure in the radiotherapy target volume suggest that patients with stage II or III non-small-cell lung cancer (NSCLC) should receive an increased total dose of radiotherapy. 2-Deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) and [18F]fluoromisonidazole ([18F]FMISO) (hypoxia) uptake on pre-radiotherapy positron emission tomography (PET)/X-ray computed tomography (CT) have been independently reported to identify intratumor subvolumes at higher risk of relapse after radiotherapy. We have compared the [18F]FDG and [18F]FMISO volumes defined by PET/CT in NSCLC patients included in a prospective study. PROCEDURES Thirty-four patients with non-resectable lung cancer underwent [18F]FDG and [18F]FMISO PET/CT before (pre-RT) and during radiotherapy (around 42 Gy, per-RT). The criteria were to delineate 40 % and 90 % SUVmax thresholds on [18F]FDG PET/CT (metabolic volumes), and SUV > 1.4 on pre-RT [18F]FMISO PET/CT (hypoxic volume). The functional volumes were delineated within the tumor volume as defined on co-registered CTs. RESULTS The mean pre-RT and per-RT [18F]FDG volumes were not statistically different (30.4 cc vs 22.2; P = 0.12). The mean pre-RT SUVmax [18F]FDG was higher than per-RT SUVmax (12.7 vs 6.5; P < 0.0001). The mean [18F]FMISO SUVmax and volumes were 2.7 and 1.37 cc, respectively. Volume-based analysis showed good overlap between [18F]FDG and [18F]FMISO for all methods of segmentation but a poor correlation for Jaccard or Dice Indices (DI). The DI maximum was 0.45 for a threshold at 40 or 50 %. CONCLUSION The correlation between [18F]FDG and [18F]FMISO uptake is low in NSCLC, making it possible to envisage different management strategies as the studies in progress show.
Collapse
Affiliation(s)
- Sébastien Thureau
- Department of Radiation Oncology, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108, FR CNRS 3638], Faculty of Medecine, University of Rouen, Rouen, France. .,Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France.
| | - R Modzelewski
- Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France
| | - P Bohn
- Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France
| | - S Hapdey
- Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France
| | - P Gouel
- Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France
| | - B Dubray
- Department of Radiation Oncology, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108, FR CNRS 3638], Faculty of Medecine, University of Rouen, Rouen, France
| | - P Vera
- Department of Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF - LITIS [EA (Equipe d'Accueil) 4108 - FR CNRS 3638], Faculty of Medicine, University of Rouen, Rouen, France
| |
Collapse
|
19
|
Zeng J, Bowen SR. Treatment Intensification in Locally Advanced/Unresectable NSCLC Through Combined Modality Treatment and Precision Dose Escalation. Semin Radiat Oncol 2021; 31:105-111. [PMID: 33610266 DOI: 10.1016/j.semradonc.2020.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The best survival for patients with unresectable, locally advanced NSCLC is currently achieved through concurrent chemoradiation followed by durvalumab for a year. Despite the best standard of care treatment, the majority of patients still develop disease recurrence, which could be distant and/or local. Trials continue to try and improve outcomes for patients with unresectable NSCLC, typically through treatment intensification, with the addition of more systemic agents, or more radiation dose to the tumor. Although RTOG 0617 showed that uniform dose escalation across an unselected population of patients undergoing chemoradiation is not beneficial, efforts continue to select patients and tumor subsets that are likely to benefit from dose escalation. This review describes some of the ongoing therapeutic trials in unresectable NSCLC, with an emphasis on quantitative imaging and precision radiation dose escalation.
Collapse
Affiliation(s)
- Jing Zeng
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA.
| | - Stephen R Bowen
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA; Department of Radiology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
20
|
Castellano A, Bailo M, Cicone F, Carideo L, Quartuccio N, Mortini P, Falini A, Cascini GL, Minniti G. Advanced Imaging Techniques for Radiotherapy Planning of Gliomas. Cancers (Basel) 2021; 13:cancers13051063. [PMID: 33802292 PMCID: PMC7959155 DOI: 10.3390/cancers13051063] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 02/07/2023] Open
Abstract
The accuracy of target delineation in radiation treatment (RT) planning of cerebral gliomas is crucial to achieve high tumor control, while minimizing treatment-related toxicity. Conventional magnetic resonance imaging (MRI), including contrast-enhanced T1-weighted and fluid-attenuated inversion recovery (FLAIR) sequences, represents the current standard imaging modality for target volume delineation of gliomas. However, conventional sequences have limited capability to discriminate treatment-related changes from viable tumors, owing to the low specificity of increased blood-brain barrier permeability and peritumoral edema. Advanced physiology-based MRI techniques, such as MR spectroscopy, diffusion MRI and perfusion MRI, have been developed for the biological characterization of gliomas and may circumvent these limitations, providing additional metabolic, structural, and hemodynamic information for treatment planning and monitoring. Radionuclide imaging techniques, such as positron emission tomography (PET) with amino acid radiopharmaceuticals, are also increasingly used in the workup of primary brain tumors, and their integration in RT planning is being evaluated in specialized centers. This review focuses on the basic principles and clinical results of advanced MRI and PET imaging techniques that have promise as a complement to RT planning of gliomas.
Collapse
Affiliation(s)
- Antonella Castellano
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Michele Bailo
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Francesco Cicone
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
- Correspondence: ; Tel.: +39-0-961-369-4155
| | - Luciano Carideo
- National Cancer Institute, G. Pascale Foundation, 80131 Naples, Italy;
| | - Natale Quartuccio
- A.R.N.A.S. Ospedale Civico Di Cristina Benfratelli, 90144 Palermo, Italy;
| | - Pietro Mortini
- Department of Neurosurgery and Gamma Knife Radiosurgery, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (M.B.); (P.M.)
| | - Andrea Falini
- Neuroradiology Unit, IRCCS Ospedale San Raffaele and Vita-Salute San Raffaele University, 20132 Milan, Italy; (A.C.); (A.F.)
| | - Giuseppe Lucio Cascini
- Department of Experimental and Clinical Medicine, “Magna Graecia” University of Catanzaro, and Nuclear Medicine Unit, University Hospital “Mater Domini”, 88100 Catanzaro, Italy;
| | - Giuseppe Minniti
- Radiation Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Policlinico Le Scotte, 53100 Siena, Italy;
- IRCCS Neuromed, 86077 Pozzilli (IS), Italy
| |
Collapse
|
21
|
Mayer P, Kraft A, Witzel HR, Marnet N, Hörner N, Roth W, Heinrich S, Hackert T, Bergmann F, Kauczor HU, Klauss M, Gaida MM. Restricted Water Diffusion in Diffusion-Weighted Magnetic Resonance Imaging in Pancreatic Cancer is Associated with Tumor Hypoxia. Cancers (Basel) 2020; 13:cancers13010089. [PMID: 33396818 PMCID: PMC7801953 DOI: 10.3390/cancers13010089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is characterized by a dense network of connective tissue surrounding clusters of cancer cells, the so-called stroma. This ubiquitous connective tissue impairs the delivery of oxygen to cancer cells. This results in hypoxia, which renders the cancer more aggressive and more resistant to treatment. In the present study, we investigated whether the extent of hypoxia in pancreatic cancer can be predicted by magnetic resonance imaging (MRI), a widely used medical imaging technique. More specifically, we used an MRI sequence which can quantitate the random motion (i.e., diffusion) of water molecules within the cancer tissue, namely diffusion-weighted (DW) MRI. We found that the random motion of water molecules is lower in cancer lesions with high hypoxia compared to those with low hypoxia. The findings from our study imply that DW-MRI can be used to identify pancreatic cancer lesions with high hypoxia which are at high risk for treatment failure. Abstract Hypoxia is a hallmark of pancreatic cancer (PDAC) due to its compact and extensive fibrotic tumor stroma. Hypoxia contributes to high lethality of this disease, by inducing a more malignant phenotype and resistance to radiation and chemotherapy. Thus, non-invasive methods to quantify hypoxia could be helpful for treatment decisions, for monitoring, especially in non-resectable tumors, or to optimize personalized therapy. In the present study, we investigated whether tumor hypoxia in PDAC is reflected by diffusion-weighted magnetic resonance imaging (DW-MRI), a functional imaging technique, frequently used in clinical practice for identification and characterization of pancreatic lesions. DW-MRI assesses the tissue microarchitecture by measuring the diffusion of water molecules, which is more restricted in highly compact tissues. As reliable surrogate markers for hypoxia, we determined Blimp-1 (B-lymphocyte induced maturation protein), a transcription factor, as well as vascular endothelial growth factor (VEGF), which are up-regulated in response to hypoxia. In 42 PDAC patients, we observed a close association between restricted water diffusion in DW-MRI and tumor hypoxia in matched samples, as expressed by high levels of Blimp-1 and VEGF in tissue samples of the respective patients. In summary, our data show that DW-MRI is well suited for the evaluation of tumor hypoxia in PDAC and could potentially be used for the identification of lesions with a high hypoxic fraction, which are at high risk for failure of radiochemotherapy.
Collapse
Affiliation(s)
- Philipp Mayer
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
- Correspondence: ; Tel.: +49-6221-5637-345
| | - Anne Kraft
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Hagen R. Witzel
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nicole Marnet
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nina Hörner
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Stefan Heinrich
- Department of Surgery, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany;
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
- Clinical Pathology, Klinikum Darmstadt GmbH, 64283 Darmstadt, Germany
| | - Hans-Ulrich Kauczor
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Miriam Klauss
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Matthias M. Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
- Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, JGU-Mainz and TRON, Translational Oncology at the University Medical Center, JGU-Mainz, 55131 Mainz, Germany
| |
Collapse
|
22
|
Yoon J, Kang SY, Lee KH, Cheon GJ, Oh DY. Targeting Hypoxia Using Evofosfamide and Companion Hypoxia Imaging of FMISO-PET in Advanced Biliary Tract Cancer. Cancer Res Treat 2020; 53:471-479. [PMID: 33091966 PMCID: PMC8053876 DOI: 10.4143/crt.2020.577] [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: 06/13/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Hypoxia is widely known as one of the mechanisms of chemoresistance and as an environmental condition which triggers invasion and metastasis of cancer. Evofosfamide is a hypoxia-activated prodrug of the cytotoxin bromo-isophosphoramide mustard conjugated with 2-nitroimidazole. Biliary tract cancer (BTC) is known to contain large hypoxic area. This study evaluated the efficacy and safety of evofosfamide as a second-line treatment of advanced BTC. Materials and Methods Patients received evofosfamide at a dose of 340 mg/m2 on days 1, 8, and 15 of every 28-day cycle. Primary end-point was progression-free survival (PFS) rate at 4-months (4m-PFSR). Secondary end-points included overall survival (OS), PFS, disease control rate (DCR), metabolic response by 18F-fluorodeoxyglucose positron emission tomography (PET), hypoxic parameters evaluated by 18F-fluoromisonidazole (FMISO) PET and toxicity. Results Twenty patients were treated with evofosfamide, with 16 response-evaluable patients. There was no objective response; stable disease was observed in nine patients, with a DCR of 56.25%. 4m-PFSR was 40.6%. Median PFS was 3.60 months (95% confidence interval [CI], 1.68 to 5.52). Median OS was 6.37 months (95% CI, 3.94 to 8.79). Reduction of tumor metabolic activity was observed in eight of 15 patients (53.3%). High baseline hypoxic parameters were associated with poor PFS. Change of hypoxic parameters between pretreatment and post-treatment reflected hypoxic-activated drug response. There was no treatment-related death. Conclusion Evofosfamide as second-line treatment of advanced BTC showed acceptable safety and comparable efficacy to other agents. Changes in volumetric parameters measured with FMISO PET, showing the degree of tumor hypoxia, reflected the response to evofosfamide based on the mode of action.
Collapse
Affiliation(s)
- Jeesun Yoon
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Seo Young Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Kyung-Hun Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Do-Youn Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Pozza DH, De Mello RA, Araujo RLC, Velcheti V. MicroRNAs in Lung Cancer Oncogenesis and Tumor Suppression: How it Can Improve the Clinical Practice? Curr Genomics 2020; 21:372-381. [PMID: 33093800 PMCID: PMC7536806 DOI: 10.2174/1389202921999200630144712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background Lung cancer (LC) development is a process that depends on genetic mutations. The DNA methylation, an important epigenetic modification, is associated with the expression of non-coding RNAs, such as microRNAs. MicroRNAs are particularly essential for cell physiology, since they play a critical role in tumor suppressor gene activity. Furthermore, epigenetic disruptions are the primary event in cell modification, being related to tumorigenesis. In this context, microRNAs can be a useful tool in the LC suppression, consequently improving prognosis and predicting treatment. Conclusion This manuscript reviews the main microRNAs involved in LC and its potential clinical applications to improve outcomes, such as survival and better quality of life.
Collapse
Affiliation(s)
- Daniel Humberto Pozza
- 1Departamento de Biomedicina da Faculdade de Medicina, and Faculdade de Ciências da Nutrição e Alimentação, and I3s, Universidade do Porto, Porto, Portugal; 2Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal; 3Department of Clinical & Experimental Oncology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil; 4Precision Oncology and Health Economic Group, Nine of July University, São Paulo, Brazil; 5Department of Digestive Surgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, Brazil; 6Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, Brazil; 7Thoracic Oncology Program, NYU Langone, Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Ramon Andrade De Mello
- 1Departamento de Biomedicina da Faculdade de Medicina, and Faculdade de Ciências da Nutrição e Alimentação, and I3s, Universidade do Porto, Porto, Portugal; 2Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal; 3Department of Clinical & Experimental Oncology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil; 4Precision Oncology and Health Economic Group, Nine of July University, São Paulo, Brazil; 5Department of Digestive Surgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, Brazil; 6Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, Brazil; 7Thoracic Oncology Program, NYU Langone, Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Raphael L C Araujo
- 1Departamento de Biomedicina da Faculdade de Medicina, and Faculdade de Ciências da Nutrição e Alimentação, and I3s, Universidade do Porto, Porto, Portugal; 2Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal; 3Department of Clinical & Experimental Oncology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil; 4Precision Oncology and Health Economic Group, Nine of July University, São Paulo, Brazil; 5Department of Digestive Surgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, Brazil; 6Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, Brazil; 7Thoracic Oncology Program, NYU Langone, Perlmutter Cancer Center, New York, NY, 10016, USA
| | - Vamsidhar Velcheti
- 1Departamento de Biomedicina da Faculdade de Medicina, and Faculdade de Ciências da Nutrição e Alimentação, and I3s, Universidade do Porto, Porto, Portugal; 2Algarve Biomedical Centre, Department of Biomedical Sciences and Medicine, University of Algarve, Faro, Portugal; 3Department of Clinical & Experimental Oncology, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo, Brazil; 4Precision Oncology and Health Economic Group, Nine of July University, São Paulo, Brazil; 5Department of Digestive Surgery, Escola Paulista de Medicina, Federal University of São Paulo (UNIFESP), São Paulo, Brazil; 6Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, Brazil; 7Thoracic Oncology Program, NYU Langone, Perlmutter Cancer Center, New York, NY, 10016, USA
| |
Collapse
|
24
|
Abgral R, Bourhis D, Calais J, Lucia F, Leclère JC, Salaün PY, Vera P, Schick U. Correlation between fluorodeoxyglucose hotspots on preradiotherapy PET/CT and areas of cancer local relapse: Systematic review of literature. Cancer Radiother 2020; 24:444-452. [DOI: 10.1016/j.canrad.2020.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 04/26/2020] [Indexed: 10/24/2022]
|
25
|
Radioresistant tumours: From identification to targeting. Cancer Radiother 2020; 24:699-705. [PMID: 32753241 DOI: 10.1016/j.canrad.2020.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/15/2022]
Abstract
From surviving fraction to tumour curability, definitions of tumour radioresistance may vary depending on the view angle. Yet, mechanisms of radioresistance have been identified and involve tumour-specific oncogenic signalling pathways, tumour metabolism and proliferation, tumour microenvironment/hypoxia, genomics. Correlations between tumour biology (histology) and imaging allow theragnostic approaches that use non-invasive biological imaging using tracer functionalization of tumour pathway biomarkers, imaging of hypoxia, etc. Modelling dose prescription function based on their tumour radio-resistant factor enhancement ratio, related to metabolism, proliferation, hypoxia is an area of investigation. Yet, the delivery of dose painting by numbers/voxel-based radiotherapy with low lineal energy transfer particles may be limited by the degree of modulation complexity needed to achieve the doses needed to counteract radioresistance. Higher lineal energy transfer particles or combinations of different particles, or combinations with drugs and devices such as done with radioenhancing nanoparticles may be promising.
Collapse
|
26
|
Yang WC, Hsu FM, Yang PC. Precision radiotherapy for non-small cell lung cancer. J Biomed Sci 2020; 27:82. [PMID: 32693792 PMCID: PMC7374898 DOI: 10.1186/s12929-020-00676-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Precision medicine is becoming the standard of care in anti-cancer treatment. The personalized precision management of cancer patients highly relies on the improvement of new technology in next generation sequencing and high-throughput big data processing for biological and radiographic information. Systemic precision cancer therapy has been developed for years. However, the role of precision medicine in radiotherapy has not yet been fully implemented. Emerging evidence has shown that precision radiotherapy for cancer patients is possible with recent advances in new radiotherapy technologies, panomics, radiomics and dosiomics. This review focused on the role of precision radiotherapy in non-small cell lung cancer and demonstrated the current landscape.
Collapse
Affiliation(s)
- Wen-Chi Yang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei, Taiwan.,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Feng-Ming Hsu
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Rd, Taipei, Taiwan. .,Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Pan-Chyr Yang
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan. .,Department of Internal Medicine, National Taiwan University Hospital, No.1 Sec 1, Jen-Ai Rd, Taipei, 100, Taiwan.
| |
Collapse
|
27
|
Dubash S, Inglese M, Mauri F, Kozlowski K, Trivedi P, Arshad M, Challapalli A, Barwick T, Al-Nahhas A, Stanbridge R, Lewanski C, Berry M, Bowen F, Aboagye EO. Spatial heterogeneity of radiolabeled choline positron emission tomography in tumors of patients with non-small cell lung cancer: first-in-patient evaluation of [ 18F]fluoromethyl-(1,2- 2H 4)-choline. Theranostics 2020; 10:8677-8690. [PMID: 32754271 PMCID: PMC7392021 DOI: 10.7150/thno.47298] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/19/2020] [Indexed: 12/26/2022] Open
Abstract
Purpose: The spatio-molecular distribution of choline and its metabolites in tumors is highly heterogeneous. Due to regulation of choline metabolism by hypoxic transcriptional signaling and other survival factors, we envisage that detection of such heterogeneity in patient tumors could provide the basis for advanced localized therapy. However, non-invasive methods to assess this phenomenon in patients are limited. We investigated such heterogeneity in Non-Small Cell Lung Cancer (NSCLC) with [18F]fluoromethyl-(1,2-2H4) choline ([18F]D4-FCH) and positron emission tomography/computed tomography (PET/CT). Experimental design: [18F]D4-FCH (300.5±72.9MBq [147.60-363.6MBq]) was administered intravenously to 17 newly diagnosed NSCLC patients. PET/CT scans were acquired concurrently with radioactive blood sampling to permit mathematical modelling of blood-tissue transcellular rate constants. Comparisons were made with biopsy-derived choline kinase-α (CHKα) expression and diagnostic [18F]fluorodeoxyglucose ([18F]FDG) scans. Results: Oxidation of [18F]D4-FCH to [18F]D4-fluorobetaine was suppressed (48.58±0.31% parent at 60 min) likely due to the deuterium isotope effect embodied within the design of the radiotracer. Early (5 min) and late (60 min) images showed specific uptake of tracer in all 51 lesions (tumors, lymph nodes and metastases) from 17 patients analyzed. [18F]D4-FCH-derived uptake (SUV60max) in index primary lesions (n=17) ranged between 2.87-10.13; lower than that of [18F]FDG PET [6.89-22.64]. Mathematical modelling demonstrated net irreversible uptake of [18F]D4-FCH at steady-state, and parametric mapping of the entire tumor showed large intratumorally heterogeneity in radiotracer retention, which is likely to have influenced correlations with biopsy-derived CHKα expression. Conclusions: [18F]D4-FCH is detectable in NSCLC with large intratumorally heterogeneity, which could be exploited in the future for targeting localized therapy.
Collapse
Affiliation(s)
- Suraiya Dubash
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Marianna Inglese
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Francesco Mauri
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Kasia Kozlowski
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Pritesh Trivedi
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| | - Mubarik Arshad
- Department of Surgery and Cancer, Imperial College London, United Kingdom
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | | | - Tara Barwick
- Department of Surgery and Cancer, Imperial College London, United Kingdom
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Adil Al-Nahhas
- Department of Radiology/Nuclear Medicine, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Rex Stanbridge
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Conrad Lewanski
- Department of Surgery and Cancer, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Matthew Berry
- Department of Medicine and Integrated Care, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Frances Bowen
- Department of Medicine and Integrated Care, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College London, United Kingdom
| |
Collapse
|
28
|
Wiedenmann N, Grosu AL, Büchert M, Rischke HC, Ruf J, Bielak L, Majerus L, Rühle A, Bamberg F, Baltas D, Hennig J, Mix M, Bock M, Nicolay NH. The utility of multiparametric MRI to characterize hypoxic tumor subvolumes in comparison to FMISO PET/CT. Consequences for diagnosis and chemoradiation treatment planning in head and neck cancer. Radiother Oncol 2020; 150:128-135. [PMID: 32544609 DOI: 10.1016/j.radonc.2020.06.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Hypoxia is an essential metabolic marker that determines chemo- and radiation resistance in head-and-neck squamous cell carcinoma (HNSCC) patients. Our exploratory analysis aimed to identify multiparametric MRI (mpMRI) parameters linked to hypoxia that might be used as surrogate for [18F]FMISO-PET in diagnosis and chemoradiation treatment (CRT) of HNSCC. MATERIALS AND METHODS 21 patients undergoing definitive CRT for HNSCC were prospectively imaged with serial [18F]FMISO-PET and 3 Tesla mpMRI for T1- and T2-weighted and dynamic contrast-enhanced perfusion and diffusion-weighted measurements (ktrans, ve, kep, ADC) in weeks 0, 2 and 5 and FDG-PET in week 0. [18F]FMISO-PET-derived hypoxic subvolumes (HSV) and complementary non-hypoxic subvolumes (nonHSV) were created for tumor and lymph nodes and projected on the mpMRI scans after PET/MRI co-registration. MpMRI and [18F]FMISO-PET parameters within HSVs and nonHSVs were statistically compared. RESULTS FMISO-PET-based HSVs of the primary tumors on MRI were characterized by lower ADC at all time points (p = 0.012 at baseline; p = 0.015 in week 2) and reduced interstitial space volume fraction ve and perfusion ktrans at baseline (p = 0.006, p = 0.047) compared to nonHSVs. Hypoxic lymph nodes were characterized by significantly lower ADC values at baseline (p = 0.039), but not at later time points and a reduction in ktrans-based perfusion at week 2 (p = 0.018). CONCLUSION MpMRI parameters differ significantly between hypoxic and non-hypoxic tumor regions, defined on FMISO-PET/CT as gold standard and might represent surrogate markers for tumor hypoxia. These findings suggest that mpMRI may be useful in the future as a surrogate modality for hypoxia imaging in order to personalize CRT.
Collapse
Affiliation(s)
- Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Büchert
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans C Rischke
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bielak
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Liette Majerus
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Bamberg
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Hennig
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Bock
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
29
|
Truffault B, Bourhis D, Chaput A, Calais J, Robin P, Le Pennec R, Lucia F, Leclère JC, Gujral DM, Vera P, Salaün PY, Schick U, Abgral R. Correlation Between FDG Hotspots on Pre-radiotherapy PET/CT and Areas of HNSCC Local Relapse: Impact of Treatment Position and Images Registration Method. Front Med (Lausanne) 2020; 7:218. [PMID: 32582727 PMCID: PMC7287148 DOI: 10.3389/fmed.2020.00218] [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] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/30/2020] [Indexed: 01/04/2023] Open
Abstract
Aim: Several series have already demonstrated that intratumoral subvolumes with high tracer avidity (hotspots) in 18F-flurodesoxyglucose positron-emission tomography (FDG-PET/CT) are preferential sites of local recurrence (LR) in various solid cancers after radiotherapy (RT), becoming potential targets for dose escalation. However, studies conducted on head and neck squamous cell carcinoma (HNSCC) found only a moderate overlap between pre- and post-treatment subvolumes. A limitation of these studies was that scans were not performed in RT treatment position (TP) and were coregistred using a rigid registration (RR) method. We sought to study (i) the influence of FDG-PET/CT acquisition in TP and (ii) the impact of using an elastic registration (ER) method to improve the localization of hotpots in HNSCC. Methods: Consecutive patients with HNSCC treated by RT between March 2015 and September 2017 who underwent FDG-PET/CT in TP at initial staging (PETA) and during follow-up (PETR) were prospectively included. We utilized a control group scanned in non treatment position (NTP) from our previous retrospective study. Scans were registered with both RR and ER methods. Various sub-volumes (AX; x = 30, 40, 50, 60, 70, 80, and 90%SUVmax) within the initial tumor and in the subsequent LR (RX; x = 40 and 70%SUVmax) were overlaid on the initial PET/CT for comparison [Dice, Jaccard, overlap fraction = OF, common volume/baseline volume = AXnRX/AX, common volume/recurrent volume = AXnRX/RX]. Results: Of 199 patients included, 43 (21.6%) had LR (TP = 15; NTP = 28). The overlap between A30, A40, and A50 sub-volumes on PETA and the whole metabolic volume of recurrence R40 and R70 on PETR showed moderate to good agreements (0.41–0.64) with OF and AXnRX/RX index, regardless of registration method or patient position. Comparison of registration method demonstrated OF and AXnRX/RX indices (x = 30% to 50%SUVmax) were significantly higher with ER vs. RR in NTP (p < 0.03), but not in TP. For patient position, the OF and AXnRX/RX indices were higher in TP than in NTP when RR was used with a trend toward significance, particularly for x=40%SUVmax (0.50±0.22 vs. 0.31 ± 0.13, p = 0.094). Conclusion: Our study suggested that PET/CT acquired in TP improves results in the localization of FDG hotspots in HNSCC. If TP is not possible, using an ER method is significantly more accurate than RR for overlap estimation.
Collapse
Affiliation(s)
- Blandine Truffault
- Department of Nuclear Medicine, Brest University Hospital, Brest, France
| | - David Bourhis
- Department of Nuclear Medicine, Brest University Hospital, Brest, France.,European University of Brittany, Brest, France
| | - Anne Chaput
- Department of Nuclear Medicine, Brest University Hospital, Brest, France
| | - Jeremie Calais
- Department of Medical and Molecular Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Nuclear Medicine and Radiology, Henri Becquerel Center, QuantIF (LITIS EA 4108 - FR CNRS 3638), Rouen University Hospital, Rouen, France
| | - Philippe Robin
- Department of Nuclear Medicine, Brest University Hospital, Brest, France.,European University of Brittany, Brest, France
| | - Romain Le Pennec
- Department of Nuclear Medicine, Brest University Hospital, Brest, France
| | - François Lucia
- Department of Radiotherapy, Brest University Hospital, Brest, France
| | | | - Dorothy M Gujral
- Clinical Oncology Department, Imperial College Healthcare NHS Trust, Charing Cross Hospital, Hammersmith, London, United Kingdom.,Department of Cancer and Surgery, Imperial College London, London, United Kingdom
| | - Pierre Vera
- Department of Nuclear Medicine and Radiology, Henri Becquerel Center, QuantIF (LITIS EA 4108 - FR CNRS 3638), Rouen University Hospital, Rouen, France
| | - Pierre-Yves Salaün
- Department of Nuclear Medicine, Brest University Hospital, Brest, France.,European University of Brittany, Brest, France
| | - Ulrike Schick
- Department of Radiotherapy, Brest University Hospital, Brest, France
| | - Ronan Abgral
- Department of Nuclear Medicine, Brest University Hospital, Brest, France.,European University of Brittany, Brest, France
| |
Collapse
|
30
|
Texte E, Gouel P, Thureau S, Lequesne J, Barres B, Edet-Sanson A, Decazes P, Vera P, Hapdey S. Impact of the Bayesian penalized likelihood algorithm (Q.Clear®) in comparison with the OSEM reconstruction on low contrast PET hypoxic images. EJNMMI Phys 2020; 7:28. [PMID: 32399752 PMCID: PMC7218037 DOI: 10.1186/s40658-020-00300-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 04/28/2020] [Indexed: 02/08/2023] Open
Abstract
Purpose To determine the impact of the Bayesian penalized likelihood (BPL) reconstruction algorithm in comparison to OSEM on hypoxia PET/CT images of NSCLC using 18F-MIZO and 18F-FAZA. Materials and methods Images of low-contrasted (SBR = 3) micro-spheres of Jaszczak phantom were acquired. Twenty patients with lung neoplasia were included. Each patient benefitted from 18F-MISO and/or 18F-FAZA PET/CT exams, reconstructed with OSEM and BPL. Lesion was considered as hypoxic if the lesion SUVmax > 1.4. A blind evaluation of lesion detectability and image quality was performed on a set of 78 randomized BPL and OSEM images by 10 nuclear physicians. SUVmax, SUVmean, and hypoxic volumes using 3 thresholding approaches were measured and compared for each reconstruction. Results The phantom and patient datasets showed a significant increase of quantitative parameters using BPL compared to OSEM but had no impact on detectability. The optimal beta parameter determined by the phantom analysis was β350. Regarding patient data, there was no clear trend of image quality improvement using BPL. There was no correlation between SUVmax increase with BPL and either SUV or hypoxic volume from the initial OSEM reconstruction. Hypoxic volume obtained by a SUV > 1.4 thresholding was not impacted by the BPL reconstruction parameter. Conclusion BPL allows a significant increase in quantitative parameters and contrast without significantly improving the lesion detectability or image quality. The variation in hypoxic volume by BPL depends on the method used but SUV > 1.4 thresholding seems to be the more robust method, not impacted by the reconstruction method (BPL or OSEM). Trial registration ClinicalTrials.gov, NCT02490696. Registered 1 June 2015
Collapse
Affiliation(s)
- Edgar Texte
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France
| | - Pierrick Gouel
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France.,QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France
| | - Sébastien Thureau
- QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France.,Radiotherapy Department, Henri Becquerel Cancer Center, Rouen, France
| | - Justine Lequesne
- Clinical Research Department, Henri Becquerel Cancer Center, Rouen, France
| | - Bertrand Barres
- Nuclear Medicine Department, Jean Perrin Cancer Center, Clermont-Ferrand, France
| | - Agathe Edet-Sanson
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France.,QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France
| | - Pierre Decazes
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France.,QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France
| | - Pierre Vera
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France.,QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France
| | - Sébastien Hapdey
- Nuclear Medicine Department, Henri Becquerel Cancer Center, Rouen, France. .,QuantIF-LITIS EA4108, Rouen University Hospital, Rouen, France.
| |
Collapse
|
31
|
Lucia F, Miranda O, Abgral R, Bourbonne V, Dissaux G, Pradier O, Hatt M, Schick U. Use of Baseline 18 F-FDG PET/CT to Identify Initial Sub-Volumes Associated With Local Failure After Concomitant Chemoradiotherapy in Locally Advanced Cervical Cancer. Front Oncol 2020; 10:678. [PMID: 32457839 PMCID: PMC7221149 DOI: 10.3389/fonc.2020.00678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/09/2020] [Indexed: 12/27/2022] Open
Abstract
Introduction: Locally advanced cervical cancer (CC) patients treated by chemoradiotherapy (CRT) have a significant local recurrence rate. The objective of this work was to assess the overlap between the initial high-uptake sub-volume (V1) on baseline 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) scans and the metabolic relapse (V2) sites after CRT in locally advanced CC. Methods: PET/CT performed before treatment and at relapse in 21 patients diagnosed with LACC and treated with CRT were retrospectively analyzed. CT images at the time of recurrence were registered to baseline CT using the 3D Slicer TM Expert Automated Registration module. The corresponding PET images were then registered using the corresponding transform. The fuzzy locally adaptive Bayesian (FLAB) algorithm was implemented using 3 classes (one for the background and the other two for tumor) in PET1 to simultaneously define an overall tumor volume and the sub-volume V1. In PET2, FLAB was implemented using 2 classes (one for background, one for tumor), in order to define V2. Four indices were used to determine the overlap between V1 and V2 (Dice coefficients, overlap fraction, X = (V1nV2)/V1 and Y = (V1nV2)/V2). Results: The mean (±standard deviation) follow-up was 26 ± 11 months. The measured overlaps between V1 and V2 were moderate to good according to the four metrics, with 0.62-0.81 (0.72 ± 0.05), 0.72-1.00 (0.85 ± 0.10), 0.55-1.00 (0.73 ± 0.16) and 0.50-1.00 (0.76 ± 0.12) for Dice, overlap fraction, X and Y, respectively. Conclusion: In our study, the overlaps between the initial high-uptake sub-volume and the recurrent metabolic volume showed moderate to good concordance. These results now need to be confirmed in a larger cohort using a more standardized patient repositioning procedure for sequential PET/CT imaging, as there is potential for RT dose escalation exploiting the pre-treatment PET high-uptake sub-volume.
Collapse
Affiliation(s)
- François Lucia
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Omar Miranda
- Radiation Oncology Department, University Hospital, Brest, France
| | - Ronan Abgral
- Nuclear Medicine Department, University Hospital, Brest, France
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Gurvan Dissaux
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Olivier Pradier
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| |
Collapse
|
32
|
Impact of positron emission tomography with computed tomography for image-guided radiotherapy. Cancer Radiother 2020; 24:362-367. [PMID: 32284178 DOI: 10.1016/j.canrad.2020.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/10/2020] [Accepted: 03/12/2020] [Indexed: 12/27/2022]
Abstract
Therapeutic effectiveness in radiotherapy is partly related to correct staging of the disease and then precise therapeutic targeting. Positron emission tomography (PET) allows the stage of many cancers to be determined and therefore is essential before deciding on radiation treatment. The definition of the therapeutic target is essential to obtain correct tumour control and limit side effects. The part of adaptive radiotherapy remains to be defined, but PET by its functional nature makes it possible to define the prognosis of many cancers and to consider radiotherapy adapted to the initial response allowing an increase over the entire metabolic volume, or targeted at a subvolume at risk per dose painting, or with a decrease in the dose in case of good response at interim assessment.
Collapse
|
33
|
Abrantes AM, Pires AS, Monteiro L, Teixo R, Neves AR, Tavares NT, Marques IA, Botelho MF. Tumour functional imaging by PET. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165717. [PMID: 32035103 DOI: 10.1016/j.bbadis.2020.165717] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/15/2020] [Accepted: 01/30/2020] [Indexed: 12/18/2022]
Abstract
Carcinogenesis is a complex multistep process, characterized by changes at different levels, both genetic and epigenetic, which alter cell metabolism. Positron emission tomography (PET) is a very sensitive image modality that allows to evaluate oncometabolism. PET functionalities are immense, since by labelling a molecule that specifically intervenes in a biochemical regulatory pathway of interest with a positron-emitting radionuclide, we can easily image that pathway. Thus, PET makes possible imaging several metabolic processes and assessing risk prediction, screening, diagnosis, response to therapy, metastization and recurrence. In this paper, we provide an overview of different radiopharmaceuticals developed for PET use in oncology, with a focus on brain tumours, breast cancer, hepatocellular carcinoma, neuroendocrine tumours, bladder cancer and prostate cancer because for these cancer types PET has been shown to be valuable. Most of the described tracers are just used in the research environment, with the aim to assess if these tracers could be able to offer an improvement concerning staging/restaging, characterization and stratification of different types of cancer, as well as therapeutic response assessment. In pursuit of personalized therapy, we briefly discuss the more established metabolic tracers and describe recent work on the development of new radiopharmaceuticals, aware that there will continue to exist diagnostic challenges to face modern cancer medicine.
Collapse
Affiliation(s)
- Ana Margarida Abrantes
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Ana Salomé Pires
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Lúcia Monteiro
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ricardo Teixo
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ana Rita Neves
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Project Development Office, Department of Mathematics and Computer Science, Eindhoven University of Technology (TU/e), NL-5612 AE Eindhoven, the Netherlands
| | - Nuno Tiago Tavares
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês Alexandra Marques
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Biophysics Institute, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; Clinical Academic Center of Coimbra, 3004-561 Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; CNC.IBILI Consortium/Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|
34
|
Gérard M, Corroyer-Dulmont A, Lesueur P, Collet S, Chérel M, Bourgeois M, Stefan D, Limkin EJ, Perrio C, Guillamo JS, Dubray B, Bernaudin M, Thariat J, Valable S. Hypoxia Imaging and Adaptive Radiotherapy: A State-of-the-Art Approach in the Management of Glioma. Front Med (Lausanne) 2019; 6:117. [PMID: 31249831 PMCID: PMC6582242 DOI: 10.3389/fmed.2019.00117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/13/2019] [Indexed: 01/31/2023] Open
Abstract
Severe hypoxia [oxygen partial pressure (pO2) below 5–10 mmHg] is more frequent in glioblastoma multiforme (GBM) compared to lower-grade gliomas. Seminal studies in the 1950s demonstrated that hypoxia was associated with increased resistance to low–linear energy transfer (LET) ionizing radiation. In experimental conditions, the total radiation dose has to be multiplied by a factor of 3 to achieve the same cell lethality in anoxic situations. The presence of hypoxia in human tumors is assumed to contribute to treatment failures after radiotherapy (RT) in cancer patients. Therefore, a logical way to overcome hypoxia-induced radioresistance would be to deliver substantially higher doses of RT in hypoxic volumes delineated on pre-treatment imaging as biological target volumes (BTVs). Such an approach faces various fundamental, technical, and clinical challenges. The present review addresses several technical points related to the delineation of hypoxic zones, which include: spatial accuracy, quantitative vs. relative threshold, variations of hypoxia levels during RT, and availability of hypoxia tracers. The feasibility of hypoxia imaging as an assessment tool for early tumor response to RT and for predicting long-term outcomes is discussed. Hypoxia imaging for RT dose painting is likewise examined. As for the radiation oncologist's point of view, hypoxia maps should be converted into dose-distribution objectives for RT planning. Taking into account the physics and the radiobiology of various irradiation beams, preliminary in silico studies are required to investigate the feasibility of dose escalation in terms of normal tissue tolerance before clinical trials are undertaken.
Collapse
Affiliation(s)
- Michael Gérard
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | | | - Paul Lesueur
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Solène Collet
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Radiophysics, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Michel Chérel
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Mickael Bourgeois
- Team 13-Nuclear Oncology, INSERM U1232 Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), Nantes, France
| | - Dinu Stefan
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Elaine Johanna Limkin
- Department of Radiotherapy, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Cécile Perrio
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/LDM-TEP Group, GIP Cyceron, Caen, France
| | - Jean-Sébastien Guillamo
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France.,Department of Neurology, Centre Hospitalier Universitaire de Nîmes, Nîmes, France
| | - Bernard Dubray
- Département de Radiothérapie et de Physique Médicale, Laboratoire QuantIF-LITIS [EA 4108], Centre de Lutte Contre le Cancer Henri Becquerel, Université de Normandie, Rouen, France
| | - Myriam Bernaudin
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| | - Juliette Thariat
- Department of Radiation Oncology, Centre Lutte Contre le Cancer François Baclesse, Caen, France
| | - Samuel Valable
- Normandie Université, UNICAEN, CEA, CNRS, ISTCT/CERVOxy Group, GIP Cyceron, Caen, France
| |
Collapse
|