1
|
Pini C, Kirienko M, Gelardi F, Bossi P, Rahal D, Toschi L, Ninatti G, Rodari M, Marulli G, Antunovic L, Chiti A, Voulaz E, Sollini M. Challenging the significance of SUV-based parameters in a large-scale retrospective study on lung lesions. Cancer Imaging 2024; 24:162. [PMID: 39593175 PMCID: PMC11600847 DOI: 10.1186/s40644-024-00807-3] [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] [Received: 04/22/2024] [Accepted: 11/19/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Although many well-known factors affect the maximum standardized uptake value (SUVmax), it remains the most requested and used parameter, especially among clinicians, despite other parameters, such as the standardized uptake value corrected for lean body mass and the metabolic tumor volume, being proven to be less sensitive to the same factors, more robust, and eventually more informative. This study intends to provide robust evidence regarding the diagnostic and prognostic value of SUVmax in a large cohort of subjects with suspected malignant lung nodules imaged by [18F]FDG PET/CT. MATERIALS AND METHODS We performed a retrospective analysis of patients with suspected/confirmed primary lung tumours undergoing [18F]FDG PET/CT. The sample size was 567 patients. Demographics, imaging, surgical, histological, and follow-up data were collected. SUVmax was analysed according to histology, stage, scanner, and outcome. The impact on measured values of different reconstruction protocols was assessed. All potential predictors of patients' outcome were assessed. RESULTS 91% cases were primary lung tumours. Lung benign nodules or metastases accounted for 5% and 4% of cases. Most patients presented with adenocarcinoma (70%) and stage I disease (51%); 144 patients relapsed and 55 died. SUVmax failed to effectively differentiate benign lesions from primary tumours or metastases. Stage I patients presented lower SUVmax. SUVmax significantly correlated with patient weight, injected [18F]FDG activity, and lesion size and differed between reconstructions' protocols. Survival analyses revealed no independent prognostic significance for SUVmax in progression-free after adjusting for other variables. SUVmax correlated with overall survival, disease stage and tumour histotype. CONCLUSION Our study confirms that SUVmax, though widely employed, present relevant limitations in discriminating between benign lesion and lung cancer, in classifying cancer histotypes, and in predicting patient outcomes independently. Known influencing factors significantly impact on numerical values, thus SUV values should be regarded with caution in clinical practice.
Collapse
Affiliation(s)
- Cristiano Pini
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Fabrizia Gelardi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20072 - Pieve, Emanuele, Italy.
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
| | - Paola Bossi
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Daoud Rahal
- Department of Pathology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Luca Toschi
- Medical Oncology and Haematology Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Gaia Ninatti
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcello Rodari
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Giuseppe Marulli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20072 - Pieve, Emanuele, Italy
- Thoracic Surgery Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | | | - Arturo Chiti
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Emanuele Voulaz
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20072 - Pieve, Emanuele, Italy
- Thoracic Surgery Unit, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Martina Sollini
- Nuclear Medicine, IRCCS San Raffaele Hospital, Milan, Italy
- Faculty of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
2
|
Maris L, Göker M, Debacker JM, De Man K, Van den Broeck B, Van Dorpe J, Van de Vijver K, Keereman V, Vanhove C. Method for co-registration of high-resolution specimen PET-CT with histopathology to improve insight into radiotracer distributions. EJNMMI Phys 2024; 11:85. [PMID: 39400788 PMCID: PMC11473743 DOI: 10.1186/s40658-024-00681-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 09/04/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND As the spatial resolution of positron emission tomography (PET) scanners improves, understanding of radiotracer distributions in tissues at high resolutions is important. Hence, we propose a method for co-registration of high-resolution ex vivo specimen PET images, combined with computed tomography (CT) images, and the corresponding specimen histopathology. METHODS We applied our co-registration method to breast cancer (BCa) specimens of patients who were preoperatively injected with 0.8 MBq/kg [18 F]fluorodeoxyglucose ([18F]FDG). The method has two components. First, we used an image acquisition scheme that minimises and tracks tissue deformation: (1) We acquired sub-millimetre (micro)-PET-CT images of ±2 mm-thick lamellas of the fresh specimens, enclosed in tissue cassettes. (2) We acquired micro-CT images of the same lamellas after formalin fixation to visualise tissue deformation. (3) We obtained 1 hematoxylin and eosin (H&E) stained histopathology section per lamella of which we captured a digital whole slide image (WSI). Second, we developed an automatic co-registration algorithm to improve the alignment between the micro-PET-CT images and WSIs, guided by the micro-CT of the fixated lamellas. To estimate the spatial co-registration error, we calculated the distance between corresponding microcalcifications in the micro-CTs and WSIs. The co-registered images allowed to study standardised uptake values (SUVs) of different breast tissues, as identified on the WSIs by a pathologist. RESULTS We imaged 22 BCa specimens, 13 cases of invasive carcinoma of no special type (NST), 6 of invasive lobular carcinoma (ILC), and 3 of ductal carcinoma in situ (DCIS). While the cassette framework minimised tissue deformation, the best alignment between the micro-PET-CT images and WSIs was achieved after deformable co-registration. We found an overall average co-registration error of 0.74 ± 0.17 mm between the micro-PET images and WSIs. (Pre)malignant tissue (including NST, ILC, and DCIS) generally showed higher SUVs than healthy tissue (including healthy glandular, connective, and adipose tissue). As expected, inflamed tissue and skin also showed high uptake. CONCLUSIONS We developed a method to co-register micro-PET-CT images of surgical specimens and WSIs with an accuracy comparable to the spatial resolution of the micro-PET images. While currently, we only applied this method to BCa specimens, we believe this method is applicable to a wide range of specimens and radiotracers, providing insight into distributions of (new) radiotracers in human malignancies at a sub-millimetre resolution.
Collapse
Affiliation(s)
- Luna Maris
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium.
- Clinical Department, XEOS Medical, Ghent, Belgium.
| | - Menekse Göker
- Department of Gynaecology, Ghent University Hospital, Ghent, Belgium
| | - Jens M Debacker
- Molecular Imaging and Therapy Research Group (MITH), Vrije Universiteit Brussel, Brussels, Belgium
- Department of Nuclear Medicine, UZ Brussel, Brussels, Belgium
- Department of Head and Skin, Head and Neck Surgery Research Group, Ghent University, Ghent, Belgium
| | - Kathia De Man
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Bliede Van den Broeck
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Koen Van de Vijver
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences and Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Vincent Keereman
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium
- Clinical Department, XEOS Medical, Ghent, Belgium
| | - Christian Vanhove
- Department of Electronics and Information Systems, Medical Image and Signal Processing, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| |
Collapse
|
3
|
Shima T, Taniguchi K, Inomata Y, Arima J, Lee SW. Glycolysis in gastrointestinal stromal tumor: a brief overview. Neoplasia 2024; 55:101022. [PMID: 38943997 PMCID: PMC11261875 DOI: 10.1016/j.neo.2024.101022] [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] [Received: 02/07/2024] [Accepted: 06/24/2024] [Indexed: 07/01/2024]
Abstract
Gastrointestinal stromal tumor (GIST) is the most prevalent mesenchymal tumor of the digestive tract. Its growth is primarily influenced by mutations in KIT or PDGFRA. Surgery is the primary treatment option for GIST; however, KIT inhibitors, such as imatinib, are used for inoperable cases. Resistance to imatinib is an upcoming challenge, especially because the effectiveness of alternative drugs is limited. Enhancement of the glycolysis pathway in cancer cells has been identified as a key feature in cancer. This unique metabolic activity has implications on tumor growth, prognosis, and resistance to therapy, even in GIST. Members of the glucose transporter (GLUT) family (particularly GLUT-1) play a significant role in GIST progression and response to treatment. Diagnostic imaging using 18F-fluorodeoxyglucose positron emission tomography/computed tomography, which enables visualization of glucose metabolism, can aid in GIST diagnosis and risk assessment. The interplay between glycolysis and GIST can lead to the development of various therapeutic strategies, especially those involving glycolysis-related molecules, such as hexokinase and lactate dehydrogenase. However, further research is required to understand the full spectrum of glycolysis in GIST and its therapeutic potential. Herein, we present an exhaustive overview and analysis of the role of glycolysis in GIST, especially as a therapeutic target.
Collapse
Affiliation(s)
- Takafumi Shima
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Kohei Taniguchi
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan; Center for Medical Research & Development, Division of Translational Research, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan.
| | - Yosuke Inomata
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Jun Arima
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| | - Sang-Woong Lee
- Department of General and Gastroenterological Surgery, Osaka Medical and Pharmaceutical University, Daigaku-machi, Takatsuki, Osaka 569-8686, Japan
| |
Collapse
|
4
|
Lee H, Hwang KH. Unexpected focal fluorodeoxyglucose uptake in main organs; pass through or pass by? World J Clin Cases 2024; 12:1885-1899. [PMID: 38660550 PMCID: PMC11036514 DOI: 10.12998/wjcc.v12.i11.1885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/31/2024] [Accepted: 03/21/2024] [Indexed: 04/11/2024] Open
Abstract
Since the inception of fluorine-18 fluorodeoxyglucose (F-18 FDG), positron emission tomography/computed tomography (PET/CT) utilizing F-18 FDG has become widely accepted as a valuable imaging modality in the field of oncology, with global prevalence in clinical practice. Given that a single Torso PET/CT scan encompasses the anatomical region from the skull base to the upper thigh, the detection of incidental abnormal focal hypermetabolism in areas of limited clinical interest is both feasible and not uncommon. Numerous investigations have been undertaken to delineate the distinctive features of these findings, yet the outcomes have proven inconclusive. The incongruent results of these studies present a challenge for physicians, leaving them uncertain about the appropriate course of action. This article provides a succinct overview of the characteristics of fluorodeoxyglucose, followed by a comprehensive discussion of the imaging findings and clinical significance associated with incidental focal abnormal F-18 FDG activity in several representative organs. In conclusion, while the prevalence of unrecognized malignancy varies across organs, malignancies account for a substantial proportion, ranging from approximately one-third to over half, of incidental focal uptake. In light of these rates, physicians are urged to exercise vigilance in not disregarding unexpected uptake, facilitating more assured clinical decisions, and advocating for further active evaluation.
Collapse
Affiliation(s)
- Haejun Lee
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| | - Kyung-Hoon Hwang
- Department of Nuclear Medicine, Gachon University College of Medicine, Gil Medical Center, Incheon 21565, South Korea
| |
Collapse
|
5
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
6
|
Okamoto M, Yamaguchi S, Sawaya R, Echizenya S, Ishi Y, Kaneko S, Motegi H, Toyonaga T, Hirata K, Fujimura M. Identifying G6PC3 as a Potential Key Molecule in Hypoxic Glucose Metabolism of Glioblastoma Derived from the Depiction of 18F-Fluoromisonidazole and 18F-Fluorodeoxyglucose Positron Emission Tomography. BIOMED RESEARCH INTERNATIONAL 2024; 2024:2973407. [PMID: 38449509 PMCID: PMC10917478 DOI: 10.1155/2024/2973407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 01/17/2024] [Accepted: 02/10/2024] [Indexed: 03/08/2024]
Abstract
Purpose Glioblastoma is the most aggressive primary brain tumor, characterized by its distinctive intratumoral hypoxia. Sequential preoperative examinations using fluorine-18-fluoromisonidazole (18F-FMISO) and fluorine-18-fluorodeoxyglucose (18F-FDG) positron emission tomography (PET) could depict the degree of glucose metabolism with hypoxic condition. However, molecular mechanism of glucose metabolism under hypoxia in glioblastoma has been unclear. The aim of this study was to identify the key molecules of hypoxic glucose metabolism. Methods Using surgically obtained specimens, gene expressions associated with glucose metabolism were analyzed in patients with glioblastoma (n = 33) who underwent preoperative 18F-FMISO and 18F-FDG PET to identify affected molecules according to hypoxic condition. Tumor in vivo metabolic activities were semiquantitatively evaluated by lesion-normal tissue ratio (LNR). Protein expression was confirmed by immunofluorescence staining. To evaluate prognostic value, relationship between gene expression and overall survival was explored in another independent nonoverlapping clinical cohort (n = 17) and validated by The Cancer Genome Atlas (TCGA) database (n = 167). Results Among the genes involving glucose metabolic pathway, mRNA expression of glucose-6-phosphatase 3 (G6PC3) correlated with 18F-FDG LNR (P = 0.03). In addition, G6PC3 mRNA expression in 18F-FMISO high-accumulated glioblastomas was significantly higher than that in 18F-FMISO low-accumulated glioblastomas (P < 0.01). Protein expression of G6PC3 was consistent with mRNA expression, which was confirmed by immunofluorescence analysis. These findings indicated that the G6PC3 expression might be facilitated by hypoxic condition in glioblastomas. Next, we investigated the clinical relevance of G6PC3 in terms of prognosis. Among the glioblastoma patients who received gross total resection, mRNA expressions of G6PC3 in the patients with poor prognosis (less than 1-year survival) were significantly higher than that in the patients who survive more than 3 years. Moreover, high mRNA expression of G6PC3 was associated with poor overall survival in glioblastoma, as validated by TCGA database. Conclusion G6PC3 was affluently expressed in glioblastoma tissues with coincidentally high 18F-FDG and 18F-FMISO accumulation. Further, it might work as a prognostic biomarker of glioblastoma. Therefore, G6PC3 is a potential key molecule of glucose metabolism under hypoxia in glioblastoma.
Collapse
Affiliation(s)
- Michinari Okamoto
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Shigeru Yamaguchi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Ryosuke Sawaya
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Sumire Echizenya
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Yukitomo Ishi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Sadahiro Kaneko
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Hiroaki Motegi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Takuya Toyonaga
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | - Miki Fujimura
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
7
|
Kim H, Choi SY, Heo TY, Kim KR, Lee J, Yoo MY, Lee TG, Han JH. Value of glucose transport protein 1 expression in detecting lymph node metastasis in patients with colorectal cancer. World J Clin Cases 2024; 12:931-941. [PMID: 38414613 PMCID: PMC10895641 DOI: 10.12998/wjcc.v12.i5.931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/04/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND There are limited data on the use of glucose transport protein 1 (GLUT-1) expression as a biomarker for predicting lymph node metastasis in patients with colorectal cancer. GLUT-1 and GLUT-3, hexokinase (HK)-II, and hypoxia-induced factor (HIF)-1 expressions may be useful biomarkers for detecting primary tumors and lymph node metastasis when combined with fluorodeoxyglucose (FDG) uptake on positron emission tomography/computed tomography (PET/CT). AIM To evaluate GLUT-1, GLUT-3, HK-II, and HIF-1 expressions as biomarkers for detecting primary tumors and lymph node metastasis with 18F-FDG-PET/CT. METHODS This retrospective study included 169 patients with colorectal cancer who underwent colectomy and preoperative 18F-FDG-PET/CT at Chungbuk National University Hospital between January 2009 and May 2012. Two tissue cores from the central and peripheral areas of the tumors were obtained and were examined by a dedicated pathologist, and the expressions of GLUT-1, GLUT-3, HK-II, and HIF-1 were determined using immunohistochemical staining. We analyzed the correlations among their expressions, various clinicopathological factors, and the maximum standardized uptake value (SUVmax) of PET/CT. RESULTS GLUT-1 was found at the center or periphery of the tumors in 109 (64.5%) of the 169 patients. GLUT-1 positivity was significantly correlated with the SUVmax of the primary tumor and lymph nodes, regardless of the biopsy site (tumor center, P < 0.001 and P = 0.012; tumor periphery, P = 0.030 and P = 0.010, respectively). GLUT-1 positivity and negativity were associated with higher and lower sensitivities of PET/CT, respectively, for the detection of lymph node metastasis, regardless of the biopsy site. GLUT3, HK-II, and HIF-1 expressions were not significantly correlated with the SUVmax of the primary tumor and lymph nodes. CONCLUSION GLUT-1 expression was significantly correlated with the SUVmax of 18F-FDG-PET/CT for primary tumors and lymph nodes. Clinicians should consider GLUT-1 expression in preoperative endoscopic biopsy in interpreting PET/CT findings.
Collapse
Affiliation(s)
- Hongsik Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju 28644, South Korea
| | - Song-Yi Choi
- Department of Pathology, Chungnam National University College of Medicine, Daejeon 35015, South Korea
| | - Tae-Young Heo
- Information and Statistics, Chungbuk National University, Cheongju 28644, South Korea
| | - Kyeong-Rok Kim
- Information and Statistics, Chungbuk National University, Cheongju 28644, South Korea
| | - Jisun Lee
- Department of Radiology, College of Medicine, Chungbuk National University, Chungbuk National University Hospital, Cheongju-si 28644, South Korea
| | - Min Young Yoo
- Department of Nuclear Medicine, School of Medicine, Inha University, Incheon 22332, South Korea
| | - Taek-Gu Lee
- Department of Surgery, Chungbuk National University, College of Medicine, Cheongju-si 28644, South Korea
| | - Joung-Ho Han
- Department of Internal Medicen, Chungbuk National University, College of medicine, Cheongju-si 28644, South Korea
| |
Collapse
|
8
|
Giovanella L, Campennì A, Tuncel M, Petranović Ovčariček P. Integrated Diagnostics of Thyroid Nodules. Cancers (Basel) 2024; 16:311. [PMID: 38254799 PMCID: PMC10814240 DOI: 10.3390/cancers16020311] [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: 12/23/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Thyroid nodules are common findings, particularly in iodine-deficient regions. Our paper aims to revise different diagnostic tools available in clinical thyroidology and propose their rational integration. We will elaborate on the pros and cons of thyroid ultrasound (US) and its scoring systems, thyroid scintigraphy, fine-needle aspiration cytology (FNAC), molecular imaging, and artificial intelligence (AI). Ultrasonographic scoring systems can help differentiate between benign and malignant nodules. Depending on the constellation or number of suspicious ultrasound features, a FNAC is recommended. However, hyperfunctioning thyroid nodules are presumed to exclude malignancy with a very high negative predictive value (NPV). Particularly in regions where iodine supply is low, most hyperfunctioning thyroid nodules are seen in patients with normal thyroid-stimulating hormone (TSH) levels. Thyroid scintigraphy is essential for the detection of these nodules. Among non-toxic thyroid nodules, a careful application of US risk stratification systems is pivotal to exclude inappropriate FNAC and guide the procedure on suspicious ones. However, almost one-third of cytology examinations are rendered as indeterminate, requiring "diagnostic surgery" to provide a definitive diagnosis. 99mTc-methoxy-isobutyl-isonitrile ([99mTc]Tc-MIBI) and [18F]fluoro-deoxy-glucose ([18F]FDG) molecular imaging can spare those patients from unnecessary surgeries. The clinical value of AI in the evaluation of thyroid nodules needs to be determined.
Collapse
Affiliation(s)
- Luca Giovanella
- Department of Nuclear Medicine, Gruppo Ospedaliero Moncucco SA, Clinica Moncucco, 6900 Lugano, Switzerland
- Clinic for Nuclear Medicine, University Hospital Zürich, 8004 Zürich, Switzerland
| | - Alfredo Campennì
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98100 Messina, Italy;
| | - Murat Tuncel
- Department of Nuclear Medicine, Hacettepe University, 06230 Ankara, Turkey;
| | - Petra Petranović Ovčariček
- Department of Oncology and Nuclear Medicine, University Hospital Center Sestre Milosrdnice, 10 000 Zagreb, Croatia;
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| |
Collapse
|
9
|
孔 德, 宋 丽, 向 阳. [Construction of a prognostic nomogram combining PET/CT metabolic parameters and blood inflammatory markers for non-small cell lung cancer treated with first-line chemotherapy]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:2139-2144. [PMID: 38189402 PMCID: PMC10774112 DOI: 10.12122/j.issn.1673-4254.2023.12.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Indexed: 01/09/2024]
Abstract
OBJECTIVE To investigate the prognostic value of 18F-FDG-PET/CT metabolic parameters and blood inflammatory markers for advanced non-small cell lung cancer (NSCLC, stage Ⅳ/ⅢB) treated with first-line chemotherapy combined with immunotherapy and construct a nomogram prediction model for NSCLC. METHODS We retrospectively analyzed the metabolic parameters (SUVmax, MTV and TLG) and blood markers of inflammation (NLR, DNLR, PLR and SII) in 105 patients with advanced NSCLC receiving chemotherapy combined with baseline 18F-FDG-PET/CT prior to immunotherapy from March, 2019 to June, 2021. ROC curve was used to calculate the best cut-off points for grouping, and univariate and multivariate COX regression analyses were performed to screen the independent predictors of prognosis for a combined diagnostic analysis. The effective biomarkers were included in the prediction model, and the nomogram model was constructed using the cph function in the rms function package of R language software. RESULTS The patients were followed up for a median of 17.5 months, and their median progression-free survival (PFS) was 16 months with a median overall survival (OS) of 13.6 months. A high PLR (≥151.050) and a high TLG (≥101.940) were significant independent prognostic factors for PFS, and a high SII (≥941.385) and a high TLG (≥101.940) were independent prognostic factors for OS. The nomogram combining PET and blood markers of inflammation showed a good performance for prognostic prediction (with C-index of 0.682 for PFS and of 0.727 for OS) and good fitting of the calibration curve. The clinical decision curve showed good clinical utility of the nomogram. CONCLUSION The baseline PET/CT metabolic parameters and blood inflammatory markers are associated with PFS and OS of patients with advanced NSCLC receiving first-line chemotherapy, and the constructed nomogram based on these parameters has a good performance for prognostic prediction in these patients.
Collapse
Affiliation(s)
- 德贤 孔
- />锦州医科大学附属第一医院核医学科,辽宁 锦州 121000Department of Nuclear Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - 丽萍 宋
- />锦州医科大学附属第一医院核医学科,辽宁 锦州 121000Department of Nuclear Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| | - 阳 向
- />锦州医科大学附属第一医院核医学科,辽宁 锦州 121000Department of Nuclear Medicine, First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121000, China
| |
Collapse
|
10
|
HAO L, WANG L, ZHANG M, YAN J, ZHANG F. [Construction of A Nomogram Prediction Model for PD-L1 Expression
in Non-small Cell Lung Cancer Based on 18F-FDG PET/CT Metabolic Parameters]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2023; 26:833-842. [PMID: 38061885 PMCID: PMC10714048 DOI: 10.3779/j.issn.1009-3419.2023.101.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND In recent years, immunotherapy represented by programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) immunosuppressants has greatly changed the status of non-small cell lung cancer (NSCLC) treatment. PD-L1 has become an important biomarker for screening NSCLC immunotherapy beneficiaries, but how to easily and accurately detect whether PD-L1 is expressed in NSCLC patients is a difficult problem for clinicians. The aim of this study was to construct a Nomogram prediction model of PD-L1 expression in NSCLC patients based on 18F-fluorodeoxy glucose (18F-FDG) positron emission tomography/conputed tomography (PET/CT) metabolic parameters and to evaluate its predictive value. METHODS Retrospective collection of 18F-FDG PET/CT metabolic parameters, clinicopathological information and PD-L1 test results of 155 NSCLC patients from Inner Mongolia People's Hospital between September 2016 and July 2021. The patients were divided into the training group (n=117) and the internal validation group (n=38), and another 51 cases of NSCLC patients in our hospital between August 2021 and July 2022 were collected as the external validation group according to the same criteria. Then all of them were categorized according to the results of PD-L1 assay into PD-L1+ group and PD-L1- group. The metabolic parameters and clinicopathological information of patients in the training group were analyzed by univariate and binary Logistic regression, and a Nomogram prediction model was constructed based on the screened independent influencing factors. The effect of the model was evaluated by receiver operating characteristic (ROC) curve, calibration curve and decision curve analysis (DCA) in both the training group and the internal and external validation groups. RESULTS Binary Logistic regression analysis showed that metabolic tumor volume (MTV), gender and tumor diameter were independent influences on PD-L1 expression. Then a Nomogram prediction model was constructed based on the above independent influences. The ROC curve for the model in the training group shows an area under the curve (AUC) of 0.769 (95%CI: 0.683-0.856) with an optimal cutoff value of 0.538. The AUC was 0.775 (95%CI: 0.614-0.936) in the internal validation group and 0.752 (95%CI: 0.612-0.893) in the external validation group. The calibration curves were tested by the Hosmer-Lemeshow test and showed that the training group (χ2=0.040, P=0.979), the internal validation group (χ2=2.605, P=0.271), and the external validation group (χ2=0.396, P=0.820) were well calibrated. The DCA curves show that the model provides clinical benefit to patients over a wide range of thresholds (training group: 0.00-0.72, internal validation group: 0.00-0.87, external validation group: 0.00-0.66). CONCLUSIONS The Nomogram prediction model constructed on the basis of 18F-FDG PET/CT metabolic parameters has greater application value in predicting PD-L1 expression in NSCLC patients.
Collapse
|
11
|
d’Abadie P, Gheysens O, Lhommel R, Jamar F, Kirchgesner T, Mazzeo F, Coubeau L, Yildiz H, De Roo AK, Schubert T. Diagnostic Superiority of Dual-Time Point [ 18F]FDG PET/CT to Differentiate Malignant from Benign Soft Tissue Tumors. Diagnostics (Basel) 2023; 13:3202. [PMID: 37892023 PMCID: PMC10606132 DOI: 10.3390/diagnostics13203202] [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: 09/12/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
[18F]FDG PET/CT is used in the workup of indeterminate soft tissue tumors (STTs) but lacks accuracy in the detection of malignant STTs. The aim of this study is to evaluate whether dual-time point [18F]FDG PET/CT imaging (DTPI) can be useful in this indication. In this prospective study, [18F]FDG PET/CT imaging was performed 1 h (t1) and 3 h (t2) after injection. Tumor uptake (SUVmax) was calculated at each time point to define a retention index (RI) corresponding to the variation between t1 and t2 (%). Sixty-eight patients were included, representing 20 benign and 48 malignant tumors (including 40 sarcomas). The RI was significantly higher in malignant STTs than in benign STTs (median: +21.8% vs. -2%, p < 0.001). An RI of >14.3% predicted STT malignancy with a specificity (Sp) of 90% and a sensitivity (Se) of 69%. An SUVmaxt1 of >4.5 was less accurate with an Sp of 80% and an Se of 60%. In a subgroup of tumors with at least mild [18F]FDG uptake (SUVmax ≥ 3; n = 46), the RI significantly outperformed the diagnostic accuracy of SUVmax (AUC: 0.88 vs. 0.68, p = 0.01). DTPI identifies malignant STT tumors with high specificity and outperforms the diagnostic accuracy of standard PET/CT.
Collapse
Affiliation(s)
- Philippe d’Abadie
- Department of Nuclear Medicine, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium; (O.G.); (R.L.); (F.J.)
| | - Olivier Gheysens
- Department of Nuclear Medicine, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium; (O.G.); (R.L.); (F.J.)
| | - Renaud Lhommel
- Department of Nuclear Medicine, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium; (O.G.); (R.L.); (F.J.)
| | - François Jamar
- Department of Nuclear Medicine, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium; (O.G.); (R.L.); (F.J.)
| | - Thomas Kirchgesner
- Department of Radiology, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Filomena Mazzeo
- Department of Clinical Oncology, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Laurent Coubeau
- Department of Abdominal Surgery, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Halil Yildiz
- Department of Internal Medicine, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - An-Katrien De Roo
- Department of Pathology, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| | - Thomas Schubert
- Department of Orthopedic Surgery, Cliniques Universitaires Saint Luc-Institut Roi Albert II, Université Catholique de Louvain, 1200 Brussels, Belgium;
| |
Collapse
|
12
|
Shen X, Zhou X, Yao Y, Meng X, Song Y, Yang Z, Li N. Superiority of [ 68Ga]Ga-DOTA-FAPI-04 PET/CT to [ 18F]FDG PET/CT in the evaluation of thymic epithelial tumours. Eur J Nucl Med Mol Imaging 2023; 50:3414-3424. [PMID: 37316675 DOI: 10.1007/s00259-023-06294-1] [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: 03/19/2023] [Accepted: 06/04/2023] [Indexed: 06/16/2023]
Abstract
PURPOSE The purpose of this study is to compare the ability of [68Ga]Ga-DOTA-FAPI-04 PET/CT and [18F]FDG PET/CT to stratify the malignancy and invasiveness of thymic epithelial tumours (TETs). METHODS From April 2021 to November 2022, participants with suspected TETs confirmed by histopathology or follow-up imaging were prospectively analysed. All participants underwent [18F]FDG and [68Ga]Ga-DOTA-FAPI-04 PET/CT within 1 week. Clinical characteristics, CT features, and metabolic parameters (maximum standardized uptake value [SUVmax] and tumour-to-mediastinum ratio [TMR]) of subjects with different pathological types and stages were compared. The diagnostic capacities of [18F]FDG and [68Ga]Ga-DOTA-FAPI-04 PET/CT were compared using receiver operating characteristic (ROC) curves and McNemar's test. RESULTS Fifty-seven participants were included. [68Ga]Ga-DOTA-FAPI-04 PET/CT was superior to [18F]FDG PET/CT in differentiating thymomas from thymic carcinomas (TCs) (AUC: 0.99 vs. 0.90, P = 0.02). Logistic regression revealed that SUVmax-FAPI (P = 0.04) was a significant predictive factor for TCs. SUVmax-FAPI and TMR-FAPI showed an excellent ability to differentiate low-risk thymomas (types A, AB, and B1), high-risk thymomas (types B2 and B3), and TCs (both P < 0.001). In thymomas, only SUVmax-FAPI (P < 0.001), TMR-FAPI (P < 0.001), and nonsmooth edges (P = 0.02) were significantly higher in the advanced-stage (Masaoka-Koga [MK] stage III/IV) group than in the early-stage group (MK stage I/II). Compared with [18F]FDG PET/CT, [68Ga]Ga-DOTA-FAPI-04 PET/CT showed significantly higher specificity (67% [46 of 69] vs. 93% [64 of 69], P < 0.001) in the detection of lymph node metastases and higher sensitivity (49% [19 of 39] vs. 97% [38 of 39], P < 0.001) in evaluating distant metastases. Both SUVmax-FAPI and TMR-FAPI were correlated with FAP expression (both r = 0.843, P < 0.001). CONCLUSION [68Ga]Ga-DOTA-FAPI-04 PET/CT was superior to [18F]FDG PET/CT in evaluating the World Health Organization (WHO) classification, MK staging, and metastatic status of TETs. TRIAL REGISTRATION ChiCTR2000038080, registration date 2020-09-09, https://www.chictr.org.cn/com/25/showproj.aspx?proj=61192.
Collapse
Affiliation(s)
- Xiuling Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Xin Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Yuan Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Xiangxi Meng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Yufei Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China
| | - Nan Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Beijing, 100142, China.
| |
Collapse
|
13
|
Suh HY, Choi H, Cho SW, Paeng JC, Cheon GJ, Park YJ, Kang KW. FDG uptake reflects an immune-enriched subtype of thyroid cancer: Clinical implications of imaging-based molecular characterization. Cancer Med 2023; 12:17068-17077. [PMID: 37466323 PMCID: PMC10501276 DOI: 10.1002/cam4.6350] [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: 11/22/2022] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/20/2023] Open
Abstract
INTRODUCTION Iodine and FDG uptakes have been established as methods to define the biological properties of thyroid cancer. As various cells in the tumor microenvironment (TME) affect tumor metabolism, we investigated the association between glucose metabolism in thyroid cancer and the TME using transcriptomic analyses. METHODS We used F-18 FDG PET and RNA sequencing data of thyroid cancer to find associations between TME cell types and glucose metabolism. In addition, publicly available single-cell RNA sequencing data of papillary thyroid cancer was used to investigate glucose metabolism in cell types of the TME. The correlations between the FDG uptake and biological properties of the TME, including glucose metabolism and tumor differentiation score (TDS) were evaluated. Estimation of the proportions of immune and cancer cells (EPIC) was performed. The biological properties of each cell type were also assessed in the single-cell RNA sequencing data. RESULTS FDG uptake showed a positive correlation with the enrichment score of macrophages and glycolysis activity. In single-cell RNA sequencing, immune cells had both high glucose transporters (GLUTs) and glycolysis signatures, while thyrocytes including cancer cells showed relatively low GLUTs and glycolysis signatures, suggesting that FDG uptake mainly occurred in immune cells of the TME. Moreover, the high GLUTs of myeloid cells were negatively associated with TDS. CONCLUSIONS Our findings suggest that thyroid cancer with high FDG uptake can be mediated by enriched immune cells of the TME. We suggest that FDG uptake in thyroid cancer could be a marker for the immune-rich type and provide clinical implications for treatment stratification.
Collapse
Affiliation(s)
- Hoon Young Suh
- Department of Nuclear MedicineSeoul National University HospitalSeoulRepublic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and TechnologySeoul National UniversitySeoulRepublic of Korea
| | - Hongyoon Choi
- Department of Nuclear MedicineSeoul National University HospitalSeoulRepublic of Korea
- Institute of Radiation Medicine, Medical Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
| | - Sun Wook Cho
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulRepublic of Korea
| | - Jin Chul Paeng
- Department of Nuclear MedicineSeoul National University HospitalSeoulRepublic of Korea
- Institute of Radiation Medicine, Medical Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear MedicineSeoul National University HospitalSeoulRepublic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and TechnologySeoul National UniversitySeoulRepublic of Korea
- Institute of Radiation Medicine, Medical Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
- Cancer Research InstituteSeoul National UniversitySeoulRepublic of Korea
- Institute on AgingSeoul National UniversitySeoulRepublic of Korea
| | - Young Joo Park
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and TechnologySeoul National UniversitySeoulRepublic of Korea
- Department of Internal MedicineSeoul National University HospitalSeoulRepublic of Korea
- Department of Internal MedicineSeoul National University College of MedicineSeoulRepublic of Korea
- Genomic Medicine Institute, Medical Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
| | - Keon Wook Kang
- Department of Nuclear MedicineSeoul National University HospitalSeoulRepublic of Korea
- Institute of Radiation Medicine, Medical Research CenterSeoul National University College of MedicineSeoulRepublic of Korea
- Cancer Research InstituteSeoul National UniversitySeoulRepublic of Korea
- Department of Biomedical SciencesSeoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
14
|
Lei P, Wang W, Sheldon M, Sun Y, Yao F, Ma L. Role of Glucose Metabolic Reprogramming in Breast Cancer Progression and Drug Resistance. Cancers (Basel) 2023; 15:3390. [PMID: 37444501 PMCID: PMC10341343 DOI: 10.3390/cancers15133390] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
The involvement of glucose metabolic reprogramming in breast cancer progression, metastasis, and therapy resistance has been increasingly appreciated. Studies in recent years have revealed molecular mechanisms by which glucose metabolic reprogramming regulates breast cancer. To date, despite a few metabolism-based drugs being tested in or en route to clinical trials, no drugs targeting glucose metabolism pathways have yet been approved to treat breast cancer. Here, we review the roles and mechanisms of action of glucose metabolic reprogramming in breast cancer progression and drug resistance. In addition, we summarize the currently available metabolic inhibitors targeting glucose metabolism and discuss the challenges and opportunities in targeting this pathway for breast cancer treatment.
Collapse
Affiliation(s)
- Pan Lei
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China; (P.L.); (W.W.)
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Wenzhou Wang
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China; (P.L.); (W.W.)
| | - Marisela Sheldon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Fan Yao
- Hubei Hongshan Laboratory, College of Biomedicine and Health, Huazhong Agricultural University, Wuhan 430070, China; (P.L.); (W.W.)
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen 518000, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston TX 77030, USA
| |
Collapse
|
15
|
Debacker JM, Maris L, Cordier F, Creytens D, Deron P, Descamps B, D'Asseler Y, De Man K, Keereman V, Libbrecht S, Schelfhout V, Van de Vijver K, Vanhove C, Huvenne W. Direct co-registration of [ 18F]FDG uptake and histopathology in surgically excised malignancies of the head and neck: a feasibility study. Eur J Nucl Med Mol Imaging 2023; 50:2127-2139. [PMID: 36854863 DOI: 10.1007/s00259-023-06153-z] [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] [Received: 08/18/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Recent technical advancements in PET imaging have improved sensitivity and spatial resolution. Consequently, clinical nuclear medicine will be confronted with PET images on a previously unfamiliar resolution. To better understand [18F]FDG distribution at submillimetric scale, a direct correlation of radionuclide-imaging and histopathology is required. METHODS A total of five patients diagnosed with a malignancy of the head and neck were injected with a clinical activity of [18F]FDG before undergoing surgical resection. The resected specimen was imaged using a preclinical high-resolution PET/CT, followed by slicing of the specimen. Multiple slices were rescanned using a micro-PET/CT device, and one of the slices was snap-frozen for frozen sections. Frozen sections were placed on an autoradiographic film, followed by haematoxylin and eosin staining to prepare them for histopathological assessment. The results from both autoradiography and histopathology were co-registered using an iterative co-registration algorithm, and regions of interest were identified to study radiotracer uptake. RESULTS The co-registration between the autoradiographs and their corresponding histopathology was successful in all specimens. The use of this novel methodology allowed direct comparison of autoradiography and histopathology and enabled the visualisation of uncharted heterogeneity in [18F]FDG uptake in both benign and malignant tissue. CONCLUSION We here describe a novel methodology enabling the direct co-registration of [18F]FDG autoradiography with the gold standard of histopathology in human malignant tissue. The future use of the current methodology could further increase our understanding of the distribution of radionuclides in surgically excised malignancies and hence, improve the integration of pathology and molecular imaging in a multiscale perspective. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05068687.
Collapse
Affiliation(s)
- Jens M Debacker
- Department of Head and Skin, Ghent University, Ghent, Belgium.
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium.
- Department of Nuclear Medicine, UZ Brussel, Brussels, Belgium.
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.
- Cancer Research Institute Ghent, Ghent, Belgium.
| | - Luna Maris
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- XEOS Medical, Ghent, Belgium
| | - Fleur Cordier
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
| | - David Creytens
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Philippe Deron
- Department of Head and Skin, Ghent University, Ghent, Belgium
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium
| | - Benedicte Descamps
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| | - Yves D'Asseler
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Kathia De Man
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Vincent Keereman
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- XEOS Medical, Ghent, Belgium
| | - Sasha Libbrecht
- Department of Pathology, Antwerp University Hospital, Edegem, Belgium
| | - Vanessa Schelfhout
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Medical Imaging, Nuclear Medicine, Ghent University Hospital, Ghent, Belgium
| | - Koen Van de Vijver
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Pathology, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Christian Vanhove
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Electronics and Information Systems, Ghent University, Ghent, Belgium
- INFINITY Lab, Ghent University, Ghent, Belgium
| | - Wouter Huvenne
- Department of Head and Skin, Ghent University, Ghent, Belgium
- Department of Head and Neck Surgery, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| |
Collapse
|
16
|
de Koster EJ, van Engen-van Grunsven ACH, Bussink J, Frielink C, de Geus-Oei LF, Kusters B, Peters H, Oyen WJG, Vriens D. [ 18F]FDG Uptake and Expression of Immunohistochemical Markers Related to Glycolysis, Hypoxia, and Proliferation in Indeterminate Thyroid Nodules. Mol Imaging Biol 2022; 25:483-494. [PMID: 36253663 PMCID: PMC10172288 DOI: 10.1007/s11307-022-01776-4] [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: 07/11/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE The current study explored the association between 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG) uptake and the quantitative expression of immunohistochemical markers related to glucose metabolism, hypoxia, and cell proliferation in benign and malignant thyroid nodules of indeterminate cytology. PROCEDURES Using a case-control design, 24 patients were selected from participants of a randomized controlled multicenter trial (NCT02208544) in which [18F]FDG-PET/CT and thyroid surgery were performed for Bethesda III and IV nodules. Three equally sized groups of [18F]FDG-positive malignant, [18F]FDG-positive benign, and [18F]FDG-negative benign nodules were included. Immunohistochemical staining was performed for glucose transporters (GLUT) 1, 3, and 4; hexokinases (HK) 1 and 2; hypoxia-inducible factor-1 alpha (HIF1α; monocarboxylate transporter 4 (MCT4); carbonic anhydrase IX (CA-IX); vascular endothelial growth factor (VEGF); sodium-iodide symporter (NIS); and Ki-67. Marker expression was scored using an immunoreactive score. Unsupervised cluster analysis was performed. The immunoreactive score was correlated to the maximum and peak standardized uptake values (SUVmax, SUVpeak) and SUVmax ratio (SUVmax of nodule/background SUVmax of contralateral, normal thyroid) of the [18F]FDG-PET/CT using the Spearman's rank correlation coefficient and compared between the three groups using Kruskal-Wallis tests. RESULTS The expression of GLUT1, GLUT3, HK2, and MCT4 was strongly positively correlated with the SUVmax, SUVpeak, and SUVmax ratio. The expression of GLUT1 (p = 0.009), HK2 (p = 0.02), MCT4 (p = 0.01), and VEGF (p = 0.007) was statistically significantly different between [18F]FDG-positive benign nodules, [18F]FDG-positive thyroid carcinomas, and [18F]FDG-negative benign nodules. In both [18F]FDG-positive benign nodules and [18F]FDG-positive thyroid carcinomas, the expression of GLUT1, HK2, and MCT4 was increased as compared to [18F]FDG-negative benign nodules. VEGF expression was higher in [18F]FDG-positive thyroid carcinomas as compared to [18F]FDG-negative and [18F]FDG-positive benign nodules. CONCLUSIONS Our results suggest that [18F]FDG-positive benign thyroid nodules undergo changes in protein expression similar to those in thyroid carcinomas. To expand the understanding of the metabolic changes in benign and malignant thyroid nodules, further research is required, including correlation with underlying genetic alterations.
Collapse
Affiliation(s)
- Elizabeth J de Koster
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands.
| | | | - Johan Bussink
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cathelijne Frielink
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands.,Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Hans Peters
- Department of Radiation Oncology, Radiotherapy & OncoImmunology Laboratory, Radboud University Medical Center, Nijmegen, Netherlands
| | - Wim J G Oyen
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands.,Department of Radiology and Nuclear Medicine, Rijnstate Hospital, Arnhem, the Netherlands.,Department of Biomedical Sciences and Humanitas Clinical and Research Centre, Department of Nuclear Medicine, Humanitas University, Milan, Italy
| | - Dennis Vriens
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
17
|
Utility of PET Scans in the Diagnosis and Management of Gastrointestinal Tumors. Dig Dis Sci 2022; 67:4633-4653. [PMID: 35908126 DOI: 10.1007/s10620-022-07616-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
|
18
|
Wang C, Xu R, Song J, Chen Y, Yin X, Ruze R, Xu Q. Prognostic value of glycolysis markers in pancreatic cancer: A systematic review and meta-analysis. Front Oncol 2022; 12:1004850. [PMID: 36172154 PMCID: PMC9510923 DOI: 10.3389/fonc.2022.1004850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction Previous studies have investigated the prognostic significance of glycolysis markers in pancreatic cancer; however, conclusions from these studies are still controversial. Methods PubMed, Embase, and Web of Science were systematically searched to investigate the prognostic role of glycolysis markers in pancreatic cancer up to May 2022. Pooled hazard ratios (HRs) with 95% confidence intervals (CIs) related to overall survival (OS), disease free survival (DFS), recurrence-free survival (RFS), and distant metastasis-free survival (DMFS) were calculated using the STATA 12.0 software. Results A total of 28 studies comprising 2010 patients were included in this meta-analysis. High expression of the five glycolysis markers was correlated with a poorer OS (HR = 1.72, 95% CI: 1.34-2.22), DFS (HR = 3.09, 95% CI: 1.91-5.01), RFS (HR = 1.73, 95% CI: 1.21-2.48) and DMFS (HR = 2.60, 95% CI: 1.09-6.20) in patients with pancreatic cancer. In subgroup analysis, it was shown that higher expression levels of the five glycolysis markers were related to a poorer OS in Asians (HR = 1.85, 95% CI: 1.46-2.35, P < 0.001) and Caucasians (HR = 1.97, 95% CI: 1.40-2.77, P < 0.001). Besides, analysis based on the expression levels of specific glycolysis markers demonstrated that higher expression levels of GLUT1 (HR = 2.11, 95% CI: 1.58-2.82, P < 0.001), MCT4 (HR = 2.26, 95% CI: 1.36-3.76, P = 0.002), and ENO1 (HR = 2.16, 95% CI: 1.28-3.66, P =0.004) were correlated with a poorer OS in patients with pancreatic cancer. Conclusions High expression of the five glycolysis markers are associated with poorer OS, DFS, RFS and DMFS in patients with pancreatic cancer, indicating that the glycolysis markers could be potential prognostic predictors and therapeutic targets in pancreatic cancer.
Collapse
|
19
|
PET imaging of pancreatic cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00207-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
20
|
van Lith SAM, Raavé R. Targets in nuclear medicine imaging: Past, present and future. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
21
|
Takahashi M, Yoshimura S, Takyu S, Aikou S, Okumura Y, Yagi K, Fukayama M, Momose T, Seto Y, Yamaya T. A design of forceps-type coincidence radiation detector for intraoperative LN diagnosis: clinical impact estimated from LNs data of 20 esophageal cancer patients. Ann Nucl Med 2021; 36:285-292. [PMID: 34843102 PMCID: PMC8897350 DOI: 10.1007/s12149-021-01701-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/22/2021] [Indexed: 11/24/2022]
Abstract
Purpose To reduce postoperative complications, intraoperative lymph node (LN) diagnosis with 18F-fluoro-2-deoxy-D-glucose (FDG) is expected to optimize the extent of LN dissection, leading to less invasive surgery. However, such a diagnostic device has not yet been realized. We proposed the concept of coincidence detection wherein a pair of scintillation crystals formed the head of the forceps. To estimate the clinical impact of this detector, we determined the cut-off value using FDG as a marker for intraoperative LN diagnosis in patients with esophageal cancer, the specifications needed for the detector, and its feasibility using numerical simulation. Methods We investigated the dataset including pathological diagnosis and radioactivity of 1073 LNs resected from 20 patients who underwent FDG-positron emission tomography followed by surgery for esophageal cancer on the same day. The specifications for the detector were determined assuming that it should measure 100 counts (less than 10% statistical error) or more within the intraoperative measurement time of 30 s. The detector sensitivity was estimated using GEANT4 simulation and the expected diagnostic ability was calculated. Results The cut-off value was 620 Bq for intraoperative LN diagnosis. The simulation study showed that the detector had a radiation detection sensitivity of 0.96%, which was better than the estimated specification needed for the detector. Among the 1035 non-metastatic LNs, 815 were below the cut-off value. Conclusion The forceps-type coincidence detector can provide sufficient sensitivity for intraoperative LN diagnosis. Approximately 80% of the prophylactic LN dissections in esophageal cancer can be avoided using this detector.
Collapse
Affiliation(s)
- Miwako Takahashi
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
- Department of Nuclear Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Shuntaro Yoshimura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Sodai Takyu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Susumu Aikou
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Okumura
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Koichi Yagi
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masashi Fukayama
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Toshimitsu Momose
- Department of Nuclear Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuyuki Seto
- Department of Gastrointestinal Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
22
|
Cazzato G, Colagrande A, Cimmino A, Abbatepaolo C, Bellitti E, Romita P, Lospalluti L, Foti C, Arezzo F, Loizzi V, Lettini T, Sablone S, Resta L, Cormio G, Ingravallo G, Rossi R. GLUT1, GLUT3 Expression and 18FDG-PET/CT in Human Malignant Melanoma: What Relationship Exists? New Insights and Perspectives. Cells 2021; 10:cells10113090. [PMID: 34831313 PMCID: PMC8624914 DOI: 10.3390/cells10113090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/03/2021] [Accepted: 11/07/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Malignant melanoma is the most aggressive of skin cancers and the 19th most common cancer worldwide, with an estimated age-standardized incidence rate of 2.8-3.1 per 100,000; although there have been clear advances in therapeutic treatment, the prognosis of MM patients with Breslow thickness greater than 1 mm is still quite poor today. The study of how melanoma cells manage to survive and proliferate by consuming glucose has been partially addressed in the literature, but some rather interesting results are starting to be present. METHODS A systematic review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and a search of PubMed and Web of Sciences (WoS) databases was performed until 27 September 2021 using the terms: glucose transporter 1 and 3 and GLUT1/3 in combination with each of the following: melanoma, neoplasm and immunohistochemistry. RESULTS In total, 46 records were initially identified in the literature search, of which six were duplicates. After screening for eligibility and inclusion criteria, 16 publications were ultimately included. CONCLUSIONS the results discussed regarding the role and expression of GLUT are still far from definitive, but further steps toward understanding and stopping this mechanism have, at least in part, been taken. New studies and new discoveries should lead to further clarification of some aspects since the various mechanisms of glucose uptake by neoplastic cells are not limited to the transporters of the GLUT family alone.
Collapse
Affiliation(s)
- Gerardo Cazzato
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
- Correspondence: ; Tel.: +39-3405203641
| | - Anna Colagrande
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Antonietta Cimmino
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Caterina Abbatepaolo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Emilio Bellitti
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Paolo Romita
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Lucia Lospalluti
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Caterina Foti
- Section of Dermatology, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (P.R.); (L.L.); (C.F.)
| | - Francesca Arezzo
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Vera Loizzi
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Teresa Lettini
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Sara Sablone
- Section of Legal Medicine, Department of Interdisciplinary Medicine, Bari Policlinico Hospital, University of Bari, “Aldo Moro”, 70124 Bari, Italy;
| | - Leonardo Resta
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Gennaro Cormio
- Section of Ginecology and Obstetrics, Department of Biomedical Sciences and Human Oncology, University of Bari, “Aldo Moro”, 70124 Bari, Italy; (F.A.); (V.L.); (G.C.)
| | - Giuseppe Ingravallo
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| | - Roberta Rossi
- Section of Pathology, Department of Emergency and Organ Transplantation (DETO), University of Bari, “Aldo Moro”, 70124 Bari, Italy; (A.C.); (A.C.); (C.A.); (E.B.); (T.L.); (L.R.); (G.I.); (R.R.)
| |
Collapse
|
23
|
Minamimoto R, Endo H. Cell Proliferation PET Imaging with 4DST PET/CT in Colorectal Adenocarcinoma and Adenoma. Diagnostics (Basel) 2021; 11:diagnostics11091658. [PMID: 34573999 PMCID: PMC8466967 DOI: 10.3390/diagnostics11091658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
An age of 70-year-old man was incidentally found two focal high 2-[18F]-fluoro-2-deoxy-d-glucose (FDG) uptake in the descending colon and in the sigmoid colon. We observed the feature of these two areas in the preplanned 4′-[methyl-11C]-thiothymidine (4DST) positron emission tomography (PET)/computed Tomography (CT)providing cell proliferation imaging. A mass forming high 4DST uptake in the descending colon and focal moderate 4DST uptake in the sigmoid colon was confirmed, and that were proven pathologically as adenocarcinoma and moderate to severe type tubular adenoma, respectively. This is the first report to present that colorectal adenoma can be visualized by proliferation PET imaging and the degree of uptake may enable discrimination of colorectal adenoma from adenocarcinoma, based on pathological considerations.
Collapse
Affiliation(s)
- Ryogo Minamimoto
- Division of Nuclear Medicine, Department of Radiology, National Center for Global Health and Medicine, Tokyo 1628655, Japan
- Correspondence: ; Tel.: +81-3-3202-7181; Fax: +81-3-3207-1038
| | - Hisako Endo
- Department of Clinical Pathology, Edogawa Hospital, Tokyo 1330052, Japan;
| |
Collapse
|
24
|
Reyes Marlés RH, Navarro Fernández JL, Puertas García-Sandoval JP, Santonja Medina F, Mohamed Salem L, Frutos Esteban L, Contreras Gutiérrez JF, Castellón Sánchez MI, Ruiz Merino G, Claver Valderas MA. Clinical value of baseline 18F-FDG PET/CT in soft tissue sarcomas. Eur J Hybrid Imaging 2021; 5:16. [PMID: 34476632 PMCID: PMC8413431 DOI: 10.1186/s41824-021-00110-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/24/2021] [Indexed: 01/18/2023] Open
Abstract
Background The use of 18F-FDG Positron emission tomography/Computed tomography (PET/CT) in the initial staging of many cancers is clearly established. Most soft tissue sarcoma (STS) has a high affinity for 18F-FDG, which is why 18F-FDG PET/CT has been proposed as a non-invasive method, useful in diagnosis and follow-up. The standardized uptake value values (SUV), the volume-based metabolic parameters MTV (metabolic tumor volume), and TLG (total lesion glycolysis) determine tumor viability and provide its total volume and the total activity of metabolically active tumor cells. The histological grade is the most important predictor of metastases and mortality associated with STS, and a significant relationship between the metabolic parameters of 18F-FDG PET/CT and the histological grade has been described. Methods A retrospective study was conducted on STS patients, who had histological grade according to the FNCLCC (Fédération Nationale des Centres de Lutte Contre Le Cancer) criteria, as well as a baseline PET/CT. SUV (SUVmax, SUVmean, and SUVpeak), MTV, and TLG were quantified. A T-student test was performed to establish the relationship between the metabolic biomarkers and the histological grade. Their usefulness as predictors of the histological grade was verified using receiver operator characteristic (ROC) curves. A survival function study was performed using the Kaplan–Meier method. To assess the prognostic utility of the metabolic biomarkers we use the Log-Rank method. Results The SUV values were useful to discriminate high-grade STS. We found a significant relationship between the histological grade and the SUV values. SUVmax, SUVpeak, MTV, and TLG were predictors of overall survival (OS). There were no significant differences in the OS for the SUVmean, or in the disease-free survival (DFS) for SUVmax, SUVmean, SUVpeak, MTV, and TLG. Conclusions The SUVmax, SUVmean, and SUVpeak values correlate with the HG and are useful to discriminate high-grade from low-grade STS. Patients with high SUVmax, SUVpeak, MTV, and TLG have a significantly lower OS.
Collapse
Affiliation(s)
- Rafael Hernando Reyes Marlés
- Nuclear Medicine Division (DIMEC), Hospitales Universitarios San Roque, Las Palmas de Gran Canaria, Las Palmas, Spain. .,Nuclear Medicine Department, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain.
| | | | | | - Fernando Santonja Medina
- Orthopedics and Traumatology Department, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | - Laroussi Mohamed Salem
- Nuclear Medicine Department, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | - Laura Frutos Esteban
- Nuclear Medicine Department, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | | | | | - Guadalupe Ruiz Merino
- Data Analytics Department, Instituto Murciano de Investigación Biosanitaria (IMIB) Virgen de la Arrixaca, El Palmar, Murcia, Spain
| | | |
Collapse
|
25
|
Pliszka M, Szablewski L. Glucose Transporters as a Target for Anticancer Therapy. Cancers (Basel) 2021; 13:cancers13164184. [PMID: 34439338 PMCID: PMC8394807 DOI: 10.3390/cancers13164184] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary For mammalian cells, glucose is a major source of energy. In the presence of oxygen, a complete breakdown of glucose generates 36 molecules of ATP from one molecule of glucose. Hypoxia is a hallmark of cancer; therefore, cancer cells prefer the process of glycolysis, which generates only two molecules of ATP from one molecule of glucose, and cancer cells need more molecules of glucose in comparison with normal cells. Increased uptake of glucose by cancer cells is due to increased expression of glucose transporters. However, overexpression of glucose transporters, promoting the process of carcinogenesis, and increasing aggressiveness and invasiveness of tumors, may have also a beneficial effect. For example, upregulation of glucose transporters is used in diagnostic techniques such as FDG-PET. Therapeutic inhibition of glucose transporters may be a method of treatment of cancer patients. On the other hand, upregulation of glucose transporters, which are used in radioiodine therapy, can help patients with cancers. Abstract Tumor growth causes cancer cells to become hypoxic. A hypoxic condition is a hallmark of cancer. Metabolism of cancer cells differs from metabolism of normal cells. Cancer cells prefer the process of glycolysis as a source of ATP. Process of glycolysis generates only two molecules of ATP per one molecule of glucose, whereas the complete oxidative breakdown of one molecule of glucose yields 36 molecules of ATP. Therefore, cancer cells need more molecules of glucose in comparison with normal cells. Increased uptake of glucose by these cells is due to overexpression of glucose transporters, especially GLUT1 and GLUT3, that are hypoxia responsive, as well as other glucose transport proteins. Increased expression of these carrier proteins may be used in anticancer therapy. This phenomenon is used in diagnostic techniques such as FDG-PET. It is also suggested, and there are observations, that therapeutic inhibition of glucose transporters may be a method in treatment of cancer patients. On the other hand, there are described cases, in which upregulation of glucose transporters, as, for example, NIS, which is used in radioiodine therapy, can help patients with cancer. The aim of this review is the presentation of possibilities, and how glucose transporters can be used in anticancer therapy.
Collapse
|
26
|
Zhang M, Yang J, Jiang H, Jiang H, Wang Z. Correlation between glucose metabolism parameters derived from FDG and tumor TNM stages and metastasis-associated proteins in colorectal carcinoma patients. BMC Cancer 2021; 21:258. [PMID: 33750337 PMCID: PMC7941722 DOI: 10.1186/s12885-021-07944-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 02/21/2021] [Indexed: 01/04/2023] Open
Abstract
Background The aim of this study was to investigate the relationship between multiple metabolism parameters derived from FDG and tumor TNM stages as well as tumor metastasis-associated protein of GLUT-1 and MACC1 in colorectal carcinoma (CRC). Methods Thirty-eight patients (24 males and 14 females) with primary CRC confirmed by elective surgery pathological, who also accepted 18F-FDG PET/CT scans during 2017 to 2019 were included in this study. The tumor classification of T, N and M is explained by the 7th American Joint Committee on Cancer (AJCC). 18F-FDG parameters of SUVmax, SUVmean, TLG and MTV were measured by drawing a region of interest on the primary lesions. The expression of GLUT-1 and MACC1 was quantified by immunohistochemical, and the correlation between metabolism parameters and tumor biomarkers were analyzed. Results According to our analysis, the 18F-FDG parameters of SUVmean was significantly correlated with tumor M status (P = 0.000) of primary CRC. The primary tumor lesion with higher SUVmax, TLG and MTV values prone to a high-T status (P = 0.002, 0.002 and 0.001, respectively). The high expression of GLUT-1/MACC1 weas more frequently involved with T3–4 stage and was poorly differentiated in CRC patients. Multivariate analysis found that the expression of GLUT-1 protein was correlated with SUVmax and MTV (R2 = 0.42, P = 0.013 and 0.004, respectively), moreover, the expression of MACC1 protein was correlated with TLG (R2 = 0.372, P = 0.000). Conclusion Glucose metabolism parameters derived from FDG provides a noninvasive assessment of M status and T status in CRC patients. The expression of GLUT-1 and MACC1 was associated with 18F-FDG uptake in CRC patients.
Collapse
Affiliation(s)
- Mingyu Zhang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, No.95 Yongan Road, Xicheng District, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, No.95 Yongan Road, Xicheng District, Beijing, China
| | - Hao Jiang
- Department of Radiology, Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, China
| | - Huijie Jiang
- Department of Radiology, Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, Heilongjiang Province, China.
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yongan Road, Xicheng District, Beijing, China.
| |
Collapse
|
27
|
Ha LN, Iravani A, Nhung NT, Hanh NTM, Hutomo F, Son MH. Relationship between clinicopathologic factors and FDG avidity in radioiodine-negative recurrent or metastatic differentiated thyroid carcinoma. Cancer Imaging 2021; 21:8. [PMID: 33413689 PMCID: PMC7792294 DOI: 10.1186/s40644-020-00378-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND In this study, we investigated the relationship between clinicopathologic factors, BRAFV600E mutation status and [18F] F-fluoro-2-deoxyglucose (FDG) avidity in patients with radioiodine (RAI)-negative recurrent or metastatic differentiated thyroid cancer (DTC). METHODS From 2015 to 2018 all patients with suspected recurrent or metastatic radioiodine-negative DTC patients who underwent FDG positron emission tomography/computed tomography (PET/CT) were retrospectively reviewed. Suspected lesions on FDG PET/CT were biopsied and underwent BRAFV600E mutation testing by immunohistochemistry and real-time PCR. Tumor size, recurrent versus metastatic disease, histopathologic features including classical type versus aggressive subtypes (poorly differentiated, tall cell, columnar cell, hobnail variants) and BRAFV600E mutation status were correlated with the SUVmax of highest hypermetabolic lesions on FDG PET/CT by the univariate analysis using logistic regression. RESULTS Sixty-three consecutive patients, 55 (87.3%) female, with median age of 48 (range 17-81) were included. The majority of patients had BRAFV600E mutation and classical subtype, 55/63 (87.3%) and 45/63(71.4%), respectively. Thyroglobulin at the time of suspected recurrence was 262.7 ng/ml (range 16.3-1000) and patients received a median 3 prior RAI treatments. Fifty-four patients (85.7%) had local recurrence. The majority of patients 58/63 (92.1%) had FDG-avid disease on PET/CT. On univariate analysis, tumor size aggressive histopathologic types and distant metastasis are the significant factors for predicting FDG uptake, p = 0.04, p = 0.001 and p = 0.004 respectively. Although FDG uptake of BRAFV600E bearing recurrent/metastatic RAIR DTC lesions was higher than those without the mutation, the difference did not reach statistical significance, SUVmax of 7.11 versus 4.91, respectively, p = 0.2. CONCLUSION The majority of recurrent or metastatic RAI-negative DTC have BRAFV600E mutation and detectable disease on FDG PET/CT. FDG avidity of the recurrent or metastatic RAI-negative DTC is independently associated with the aggressive histopathologic features.
Collapse
Affiliation(s)
- Le Ngoc Ha
- Department of Nuclear Medicine, 108 Central Military Hospital, 1st Tran Hung Dao, Hai Ba Trung, Ha Noi, Vietnam
| | - Amir Iravani
- Washington University School of Medicine, Mallinckrodt Institute of Radiology, St Louis, MO, USA
| | - Nguyen Thi Nhung
- Department of Nuclear Medicine, 108 Central Military Hospital, 1st Tran Hung Dao, Hai Ba Trung, Ha Noi, Vietnam
| | - Ngo Thi Minh Hanh
- Department of Pathology, 108 Central Military Hospital, Hanoi, Vietnam
| | - Febby Hutomo
- Nuclear Medicine Department, MRCCC Siloam Hospital, Jakarta, Indonesia
| | - Mai Hong Son
- Department of Nuclear Medicine, 108 Central Military Hospital, 1st Tran Hung Dao, Hai Ba Trung, Ha Noi, Vietnam.
| |
Collapse
|
28
|
One-by-One Comparison of Lymph Nodes Between 18F-FDG Uptake and Pathological Diagnosis in Esophageal Cancer. Clin Nucl Med 2020; 45:741-746. [PMID: 32796247 PMCID: PMC7469872 DOI: 10.1097/rlu.0000000000003224] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Esophagectomy with extended lymph node (LN) dissection is a standard treatment for resectable esophageal cancer to prevent recurrence, but severe, potentially life-threatening postoperative complications are still important issues. Accurate diagnosis of LN metastases would enable the decision to dissect or leave the LNs in regions with high risk of complications. Advancements in intraoperative gamma probe and radioactivity detectors have made intraoperative navigation surgery possible using a radiotracer as a marker. 18F-FDG is one such candidate markers, and the diagnostic power of FDG through counting the radioactivity close to each LN should be elucidated.
Collapse
|
29
|
Pezzuto A, D'Ascanio M, Ricci A, Pagliuca A, Carico E. Expression and role of p16 and GLUT1 in malignant diseases and lung cancer: A review. Thorac Cancer 2020; 11:3060-3070. [PMID: 32945604 PMCID: PMC7606016 DOI: 10.1111/1759-7714.13651] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/17/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Non‐small cell lung cancer (NSCLC) is the leading cause of cancer death and in most cases it is often diagnosed at an advanced stage. Many genetic and microenvironmental factors are able to modify the cell cycle inducing carcinogenesis and tumor growth. Among the metabolic and genetic factors that come into play in carcinogenesis and tumor cell differentiation and growth there are two different proteins that should be considered which are glucose transporters (GLUTs) and p16INK4 The first are glucose transporters which are strongly involved in tumor metabolism, notably accelerating cancer cell metabolism both in aerobic and anaerobic conditions. There are different subtypes of GLUT family factors of which GLUT 1 is the most important and widely expressed. By contrast, p16 is mainly a tumor‐suppressor protein that acts on cyclin‐dependent kinase favoring cell cycle arrest in the G1 phase. Our search focused on the action of the aforementioned factors.
Collapse
Affiliation(s)
- Aldo Pezzuto
- Cardiovascular-Pulmonary Science Department, Sant' Andrea Hospital-Sapienza University, Rome, Italy
| | - Michela D'Ascanio
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| | - Alberto Ricci
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| | - Alessandra Pagliuca
- Cardiovascular-Pulmonary Science Department, Sant' Andrea Hospital-Sapienza University, Rome, Italy
| | - Elisabetta Carico
- Clinical and Molecular Medicine Department, Sant' Andrea Hospital- Sapienza University, Rome, Italy
| |
Collapse
|
30
|
Moradi F, Iagaru A. The Role of Positron Emission Tomography in Pancreatic Cancer and Gallbladder Cancer. Semin Nucl Med 2020; 50:434-446. [PMID: 32768007 DOI: 10.1053/j.semnuclmed.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
18F-FDG-PET is complementary to conventional imaging in patients with clinical suspicion for exocrine pancreatic malignancies. It has similar if not superior sensitivity and specificity for detection of cancer, and when combined with contrast enhanced anatomic imaging of the abdomen, can improve diagnostic accuracy and aid in staging, assessment for resectability, radiation therapy planning, and prognostication. Various metabolic pathways affect FDG uptake in pancreatic ductal adenocarcinoma. The degree of uptake reflects histopathology, aggressiveness, metastatic potential, and metabolic profile of malignant cell and their interaction with cancer stroma. After treatment, FDG-PET is useful for detection of residual or recurrent cancer and can be used to assess and monitor response to therapy in unresectable or metastatic disease. The degree and pattern of uptake combined with other imaging features are useful in characterization of incidental pancreatic lesions and benign processes such as inflammation. Several novel PET radiopharmaceuticals have been developed to improve detection and management of pancreatic cancer. Gallbladder carcinoma is typically FDG avid and when anatomic imaging is equivocal PET can be used to assess metastatic involvement with high specificity and inform subsequent management.
Collapse
Affiliation(s)
- Farshad Moradi
- Division of Nuclear Medicine, Department of Radiology, Stanford University, Stanford, CA.
| | - Andrei Iagaru
- Division of Nuclear Medicine, Department of Radiology, Stanford University, Stanford, CA
| |
Collapse
|
31
|
Cheng NM, Hsieh CE, Fang YHD, Liao CT, Ng SH, Wang HM, Chou WC, Lin CY, Yen TC. Development and validation of a prognostic model incorporating [ 18F]FDG PET/CT radiomics for patients with minor salivary gland carcinoma. EJNMMI Res 2020; 10:74. [PMID: 32632638 PMCID: PMC7338312 DOI: 10.1186/s13550-020-00631-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 04/08/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES The aim of this study was to develop and validate a prognostic model incorporating [18F]FDG PET/CT radiomics for patients of minor salivary gland carcinoma (MSGC). METHODS We retrospectively reviewed the pretreatment [18F]FDG PET/CT images of 75 MSGC patients treated with curative intent. Using a 1.5:1 ratio, the patients were randomly divided into a training and validation group. The main outcome measurements were overall survival (OS) and relapse-free survival (RFS). All of the patients were followed up for at least 30 months or until death. Following segmentation of tumors and lymph nodes on PET images, radiomic features were extracted. The prognostic significance of PET radiomics and clinical parameters in the training group was examined using receiver operating characteristic curve analysis. Variables showing a significant impact on OS and RFS were entered into multivariable Cox regression models. Recursive partitioning analysis was subsequently implemented to devise a prognostic index, whose performance was examined in the validation group. Finally, the performance of the index was compared with clinical variables in the entire cohort and nomograms for surgically treated cases. RESULTS The training and validation groups consisted of 45 and 30 patients, respectively. The median follow-up time in the entire cohort was 59.5 months. Eighteen relapse, 19 dead, and thirteen relapse, eight dead events were found in the training and validation cohorts, respectively. In the training group, two factors were identified as independently associated with poor OS, i.e., (1) tumors with both high maximum standardized uptake value (SUVmax) and discretized intensity entropy and (2) poor performance status or N2c-N3 stage. A prognostic model based on the above factors was devised and showed significant higher concordance index (C-index) for OS than those of AJCC stage and high-risk histology (C-index: 0.83 vs. 0.65, P = 0.005; 0.83 vs. 0.54, P < 0.001, respectively). This index also demonstrated superior performance than nomogram for OS (C-index: 0.88 vs. 0.70, P = 0.017) and that for RFS (C-index: 0.87 vs. 0.72, P = 0.004). CONCLUSIONS We devised a novel prognostic model that incorporates [18F]FDG PET/CT radiomics and may help refine outcome prediction in patients with MSGC.
Collapse
Affiliation(s)
- Nai-Ming Cheng
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Department of Nuclear Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Cheng-En Hsieh
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yu-Hua Dean Fang
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chun-Ta Liao
- Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shu-Hang Ng
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Hung-Ming Wang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Wen-Chi Chou
- Division of Hematology/Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Tzu-Chen Yen
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan. .,Department of Nuclear Medicine, Xiamen Chang Gung Hospital, Xiamen, China.
| |
Collapse
|
32
|
L-Glucose: Another Path to Cancer Cells. Cancers (Basel) 2020; 12:cancers12040850. [PMID: 32244695 PMCID: PMC7225996 DOI: 10.3390/cancers12040850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 01/31/2023] Open
Abstract
Cancerous tumors comprise cells showing metabolic heterogeneity. Among numerous efforts to understand this property, little attention has been paid to the possibility that cancer cells take up and utilize otherwise unusable substrates as fuel. Here we discuss this issue by focusing on l-glucose, the mirror image isomer of naturally occurring d-glucose; l-glucose is an unmetabolizable sugar except in some bacteria. By combining relatively small fluorophores with l-glucose, we generated fluorescence-emitting l-glucose tracers (fLGs). To our surprise, 2-NBDLG, one of these fLGs, which we thought to be merely a control substrate for the fluorescent d-glucose tracer 2-NBDG, was specifically taken up into tumor cell aggregates (spheroids) that exhibited nuclear heterogeneity, a major cytological feature of malignancy in cancer diagnosis. Changes in mitochondrial activity were also associated with the spheroids taking up fLG. To better understand these phenomena, we review here the Warburg effect as well as key studies regarding glucose uptake. We also discuss tumor heterogeneity involving aberrant uptake of glucose and mitochondrial changes based on the data obtained by fLG. We then consider the use of fLGs as novel markers for visualization and characterization of malignant tumor cells.
Collapse
|
33
|
Lee HL, Farrell R, Kamath V, Ho-Shon I, Yap F. Concordant PET/CT and ICG positive lymph nodes in endometrial cancer: a case of mistaken identity. J Surg Case Rep 2020; 2020:rjz377. [PMID: 31976060 PMCID: PMC6963166 DOI: 10.1093/jscr/rjz377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/02/2019] [Indexed: 11/12/2022] Open
Abstract
Endometrial carcinoma is the most common gynecological malignancy in developed countries. In early stage endometrial cancer, routine systemic pelvic lymphadenectomy showed no survival benefits and results in increased morbidity. The role of PET/CT imaging for the pre-operative detection of lymph node metastases in endometrial cancer is unclear. Sentinel lymph node (SLN) mapping may reduce the surgical staging morbidity while maintaining prognostic information of the lymph node status. Recently, indocyanine green (ICG) SLN mapping has been utilized to detect nodal metastasis in endometrial cancer. Endosalpingiosis is defined as the presence of tubal-like epithelium outside of the fallopian tube and can sometimes be misinterpreted as cancer metastasis. Here, we discuss a patient with newly diagnosed endometrial cancer who had pelvic and para-aortic lymph nodes with high glucose avidity on PET/CT, and ICG positivity on SLN mapping, suspected clinically to be metastatic adenocarcinoma, but with the pathological finding of endosalpingiosis only.
Collapse
Affiliation(s)
- Hong L Lee
- Department of Gynaecology, Joondalup Health Campus, Joondalup, WA, Australia
| | - Rhonda Farrell
- Department of Gynaecological Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | - Vasanth Kamath
- Department of Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Ivan Ho-Shon
- Department of Nuclear Medicine and PET, Prince of Wales Hospital, Randwick, NSW, Australia
| | - Francis Yap
- PathWest, Department of Health, Perth,WA, Australia
| |
Collapse
|
34
|
Suh HY, Choi H, Paeng JC, Cheon GJ, Chung JK, Kang KW. Comprehensive gene expression analysis for exploring the association between glucose metabolism and differentiation of thyroid cancer. BMC Cancer 2019; 19:1260. [PMID: 31888560 PMCID: PMC6937781 DOI: 10.1186/s12885-019-6482-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/18/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The principle of loss of iodine uptake and increased glucose metabolism according to dedifferentiation of thyroid cancer is clinically assessed by imaging. Though these biological properties are widely applied to appropriate iodine therapy, the understanding of the genomic background of this principle is still lacking. We investigated the association between glucose metabolism and differentiation in advanced thyroid cancer as well as papillary thyroid cancer (PTC). METHODS We used RNA sequencing of 505 patients with PTC obtained from the Cancer Genome Archives and microarray data of poorly-differentiated and anaplastic thyroid cancer (PDTC/ATC). The signatures of GLUT and glycolysis were estimated to assess glucose metabolic profiles. The glucose metabolic profiles were associated with tumor differentiation score (TDS) and BRAFV600E mutation status. In addition, survival analysis of glucose metabolic profiles was performed for predicting recurrence-free survival. RESULTS In PTC, the glycolysis signature was positively correlated with TDS, while the GLUT signature was inversely correlated with TDS. These correlations were significantly stronger in the BRAFV600E negative group than the positive group. Meanwhile, both GLUT and glycolysis signatures were negatively correlated with TDS in advanced thyroid cancer. The high glycolysis signature was significantly associated with poor prognosis in PTC in spite of high TDS. The glucose metabolic profiles are intricately associated with tumor differentiation in PTC and PDTC/ATC. CONCLUSIONS As glycolysis was an independent prognostic marker, we suggest that the glucose metabolism features of thyroid cancer could be another biological progression marker different from differentiation and provide clinical implications for risk stratification. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hoon Young Suh
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea.
| | - Jin Chul Paeng
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - Gi Jeong Cheon
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea
- Department of Nuclear Medicine, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10408, Republic of Korea
| | - Keon Wook Kang
- Department of Nuclear Medicine, Seoul National University Hospital, 28 Yongon-Dong, Jongno-Gu, Seoul, 03080, Republic of Korea
| |
Collapse
|