1
|
Yokoyama K, Tsuchiya J, Kuwahara H, Inaji M, Tateishi U. 18 F-FAZA, 18 F-FDG, and 123 I-IMP Imaging Reveal Hypoxia and Metabolism in Acute MOGAD. Clin Nucl Med 2025; 50:e236-e237. [PMID: 39780349 PMCID: PMC11878587 DOI: 10.1097/rlu.0000000000005671] [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: 07/17/2024] [Revised: 11/08/2024] [Indexed: 01/11/2025]
Abstract
ABSTRACT We report the first case of myelin oligodendrocyte glycoprotein antibody-associated disease (MOGAD) evaluated with hypoxic imaging using 18 F-FAZA PET/CT. A healthy woman in her 20s presented to our hospital with seizures, headaches, and vomiting. MRI and CT scans suggested a wide range of differential diagnoses, from neoplastic lesions, such as malignant lymphoma, to inflammatory diseases, such as vasculitis, making her case challenging to diagnose. MOGAD was confirmed by positive myelin oligodendrocyte glycoprotein antibodies, and her condition improved with steroid therapy. The 18 F-FAZA PET/CT findings in this case may aid in understanding the pathogenesis of MOGAD.
Collapse
Affiliation(s)
| | | | | | - Motoki Inaji
- Neurosurgery, Institute of Science Tokyo, Tokyo, Japan
| | | |
Collapse
|
2
|
Alenezi A, Alhamad H, Alenezi A, Khan MU. Hypoxia Imaging in Lung Cancer: A PET-Based Narrative Review for Clinicians and Researchers. Pharmaceuticals (Basel) 2025; 18:459. [PMID: 40283896 PMCID: PMC12030053 DOI: 10.3390/ph18040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Hypoxia plays a critical role in lung cancer progression and treatment resistance by contributing to aggressive tumor behavior and poor therapeutic response. Molecular imaging, particularly positron emission tomography (PET), has become an essential tool for noninvasive hypoxia detection, providing valuable insights into tumor biology and aiding in personalized treatment strategies. Objective: This narrative review explores recent advancements in PET imaging for detecting hypoxia in lung cancer, with a focus on the development, characteristics, and clinical applications of various radiotracers. Findings: Numerous PET-based hypoxia radiotracers have been investigated, each with distinct pharmacokinetics and imaging capabilities. Established tracers such as 18F-Fluoromisonidazole (18F-FMISO) remain widely used, while newer alternatives like 18F-Fluoroazomycin Arabinoside (18F-FAZA) and 18F-Flortanidazole (18F-HX4) demonstrate improved clearance and image contrast. Additionally, 64Cu-ATSM has gained attention for its rapid tumor uptake and hypoxia selectivity. The integration of PET with hybrid imaging modalities, such as PET/CT and PET/MRI, enhances the spatial resolution and functional interpretation, making hypoxia imaging a promising approach for guiding radiotherapy, chemotherapy, and targeted therapies. Conclusions: PET imaging of hypoxia offers significant potential in lung cancer diagnosis, treatment planning, and therapeutic response assessment. However, challenges remain, including tracer specificity, quantification variability, and standardization of imaging protocols. Future research should focus on developing next-generation radiotracers with enhanced specificity, optimizing imaging methodologies, and leveraging multimodal approaches to improve clinical utility and patient outcomes.
Collapse
Affiliation(s)
- Ahmad Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Hamad Alhamad
- Occupational Therapy Department, Kuwait University, Jabriya 31470, Kuwait
| | - Aishah Alenezi
- Radiologic Sciences Department, Kuwait University, Kuwait City 31470, Kuwait
| | - Muhammad Umar Khan
- Nuclear Medicine Department, Jahra Hospital, Ministry of Health, Al Jahra 03200, Kuwait
| |
Collapse
|
3
|
Kristensen MH, Nielsen SB, Alsner J, Holm AIS, Hansen CR, Overgaard J, Eriksen JG. A systematic review and proportional meta-analysis of image-based pattern of loco-regional failure analyses outcomes in head and neck squamous cell carcinoma. Radiother Oncol 2025; 207:110838. [PMID: 40089161 DOI: 10.1016/j.radonc.2025.110838] [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: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND AND PURPOSE The prognosis following loco-regional failure after primary radiotherapy (RT) for head and neck squamous cell carcinoma (HNSCC) is poor. The hypothesis that most failures occur as a consequence of tumor radioresistance, can be evaluated by proxy as the proportion of failures that occur in the high-dose region. Several studies have investigated possible reasons for treatment failure by an image-based pattern of failure analyses (POF), comparing the initial planning CT scan with a scan conducted upon failure. The aim of the present systematic review and meta-analysis was to evaluate the proportion of failures that occurred in the high-dose region of all analyzed failures. MATERIALS AND METHODS A systematic database search from 2000 to 2023, was performed for studies including results from image-based loco-regional POF, regardless of the method, after primary RT for HNSCC. Proportions of volumetrically in-field (opposed to marginal or outfield) failures, point of origin-based inside high-dose targets, or covered by curative doses for both the number of patients and the number of failure sites were analyzed in proportional meta-analyses. The review was registered at Prospero (CRD42023412545). RESULTS Out of 56 included studies, accumulated image-based POF results were available from 1,161 patients and 658 individual failure sites. The majority of patients had in-field failures in volumetric-based studies (84 % (95 % CI: 77;90)), inside failures in point of origin-based studies (82 % (95% CI:61;85)) or failures covered by 95 % of dose prescribed to CTV1 (84 % (95% CI:69;95)). A trend toward increasing proportions of non-high-dose failures in more recently treated patients was observed. CONCLUSION Most loco-regional failures for patients treated with primary RT for HNSCC are related to the high-dose volume. Therefore, a focus on biomarkers predicting individual tumor radiosensitivity is warranted to enable individualized treatment intensification to increase loco-regional control.
Collapse
Affiliation(s)
| | - Signe Bergliot Nielsen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark; Department of Oto-Rhino-Laryngology, Head and Neck, Aarhus University Hospital, Denmark
| | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Christian Rønn Hansen
- Odense University Hospital, Laboratory of Radiation Physics, Odense, Denmark; University of Southern Denmark, Department of Clinical Research, Odense, Denmark; Danish Centre for Particle Therapy, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jesper Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
4
|
Winter RM, Boeke S, Leibfarth S, Habrich J, Clasen K, Nikolaou K, Zips D, Thorwarth D. Clinical validation of a prognostic preclinical magnetic resonance imaging biomarker for radiotherapy outcome in head-and-neck cancer. Radiother Oncol 2025; 204:110702. [PMID: 39733969 DOI: 10.1016/j.radonc.2024.110702] [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: 03/26/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/31/2024]
Abstract
PURPOSE To retrain a model based on a previously identified prognostic imaging biomarker using apparent diffusion coefficient (ADC) values from diffusion-weighted magnetic resonance imaging (DW-MRI) in a preclinical setting and validate the model using clinical DW-MRI data of patients with locally advanced head-and-neck cancer (HNC) acquired before radiochemotherapy. MATERIAL AND METHODS A total of 31 HNC patients underwent T2-weighted and DW-MRI using 3 T MRI before radiochemotherapy (35 x 2 Gy). Gross tumor volumes (GTV) were delineated based on T2-weighted and b500 images. A preclinical model previously revealed that the size of high-risk subvolumes (HRS) defined by a band of ADC-values was correlated to radiation resistance. To validate this model, different bands of ADC-values were tested using two-sided thresholds on the low-ADC histogram flank to determine HRSs inside the GTV and correlated to treatment outcome after three years. The best model was used to fit a logistic regression model. Stratification potential regarding outcome was internally validated using bootstrap, receiver-operator-characteristic (ROC)-analysis, Kaplan-Meier- and Cox-method, and compared to GTV, ADCmean and clinical factors. RESULTS The best model was defined by 800 CONCLUSIONS A preclinical prognostic model defined by an ADC-based HRS was successfully retrained and validated in HNC patients treated with radiochemotherapy. After thorough external validation, such functional HRS based on a band of ADC values may in the future allow interventional response-adaptive MRI-guided radiotherapy in online and offline approaches.
Collapse
Affiliation(s)
- René M Winter
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Simon Boeke
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Leibfarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Jonas Habrich
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Kerstin Clasen
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Konstantin Nikolaou
- Diagnostic and Interventional Radiology, Department of Radiology, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Radiation Oncology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität Zu Berlin, Berlin, Germany
| | - Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany; Cluster of Excellence "Machine Learning", University of Tübingen, Tübingen, Germany.
| |
Collapse
|
5
|
Menegakis A, Vennin C, Ient J, Groot AJ, Krenning L, Klompmaker R, Friskes A, Ilic M, Yaromina A, Harkes R, van den Broek B, Jakob Sonke J, De Jong M, Piepers J, van Rheenen J, Vooijs MA, Medema RH. A novel lineage-tracing tool reveals that hypoxic tumor cells drive tumor relapse after radiotherapy. Radiother Oncol 2025; 202:110592. [PMID: 39427933 PMCID: PMC11718160 DOI: 10.1016/j.radonc.2024.110592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024]
Abstract
PURPOSE Tumor hypoxia imposes a main obstacle to the efficacy of anti-cancer therapy. Understanding the cellular dynamics of individual hypoxic cells before, during and post-treatment has been hampered by the technical inability to identify and trace these cells over time. METHODS AND MATERIALS Here, we present a novel lineage-tracing reporter for hypoxic cells based on the conditional expression of a HIF1a-CreERT2-UnaG biosensor that can visualize hypoxic cells in a time-dependent manner and trace the fate of hypoxic cells over time. We combine this system with multiphoton microscopy, flow cytometry, and immunofluorescence to characterize the role of hypoxic cells in tumor relapse after irradiation in H1299 tumor spheroids and in vivo xenografts. RESULTS We validate the reporter in monolayer cultures and we show that tagged cells colocalize in spheroids and human tumor xenografts with the hypoxic marker pimonidazole. We found that irradiation of H1299-HIFcreUnaG spheroids leads to preferential outgrowth of cells from the hypoxic core. Similarly, in xenografts tumors, although initially UnaG-positive-cells coincide with pimonidazole-positive tumor areas and they are merely quiescent, upon irradiation UnaG-positive cells enrich in regrowing tumors and are mainly proliferative. CONCLUSIONS Collectively, our data provide clear evidence that the hypoxic cells drive tumor relapse after irradiation.
Collapse
Affiliation(s)
- Apostolos Menegakis
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland; Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands; Oncode Institute, Division of Tumor Biology and Tumor Immunology, the Netherlands.
| | - Claire Vennin
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jonathan Ient
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Arjan J Groot
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Lenno Krenning
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Rob Klompmaker
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Anoek Friskes
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Mila Ilic
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland
| | - Ala Yaromina
- The M-Lab, Department of Precision Medicine, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, the Netherlands
| | - Rolf Harkes
- Bioimaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Bram van den Broek
- Bioimaging Facility, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jan Jakob Sonke
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Monique De Jong
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Jolanda Piepers
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands
| | - Jacco van Rheenen
- Division of Molecular Pathology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Marc A Vooijs
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, 6200 MD Maastricht, the Netherlands.
| | - René H Medema
- Oncode Institute, Division of Cell Biology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherland; Princess Maxima Center for Pediatric Oncology, Utrecht, the Netherlands(2).
| |
Collapse
|
6
|
Dubec MJ, Price J, Berks M, Gaffney J, Little RA, Porta N, Sridharan N, Datta A, McHugh DJ, Hague CJ, Cheung S, Manoharan P, van Herk M, Choudhury A, Matthews JC, Parker GJ, Buckley DL, Harrington KJ, McPartlin A, O’Connor JP. Oxygen-Enhanced MRI Detects Incidence, Onset, and Heterogeneity of Radiation-Induced Hypoxia Modification in HPV-Associated Oropharyngeal Cancer. Clin Cancer Res 2024; 30:5620-5629. [PMID: 39142654 PMCID: PMC11654720 DOI: 10.1158/1078-0432.ccr-24-1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/23/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Hypoxia mediates treatment resistance in solid tumors. We evaluated if oxygen-enhanced MRI-derived hypoxic volume (HVMRI) is repeatable and can detect radiotherapy-induced hypoxia modification in human papillomavirus-associated oropharyngeal head and neck squamous cell cancer. EXPERIMENTAL DESIGN A total of 27 patients were recruited prospectively between March 2021 and January 2024. HVMRI was measured in primary and nodal tumors prior to standard-of-care (chemo)radiotherapy and then at weeks 2 and 4 (W2 and W4) into therapy. Two pretreatment scans assessed biomarker within-subject coefficient of variation and repeatability coefficient (RC). Cohort treatment response was measured using mixed-effects modeling. Responding lesions were identified by comparing HVMRI change with RC limits of agreement. RESULTS Oxygen-enhanced MRI identified hypoxia in all lesions. The HVMRI within-subject coefficient of variation was 24.6%, and RC limits of agreement were -45.7% to 84.1%. A cohort median pretreatment HVMRI of 11.3 cm3 reduced to 6.9 cm3 at W2 and 5.9 cm3 at W4 (both P < 0.001). HVMRI was reduced in 54.5% of individual lesions by W2 and in 88.2% by W4. All lesions with W2 hypoxia reduction showed persistent modification at W4. HVMRI reduced in some lesions that showed no overall volume change. Hypoxia modification was discordant between primary and nodal tumors in 50.0% of patients. CONCLUSIONS Radiation-induced hypoxia modification can occur as early as W2, but onset varies between patients and was not necessarily associated with overall size change. Half of all patients had discordant changes in primary and nodal tumors. These findings have implications for patient selection and timing of dose de-escalation strategies in human papillomavirus-associated oropharyngeal carcinoma. See related commentary by Mason, p. 5503.
Collapse
Affiliation(s)
- Michael J. Dubec
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - James Price
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Michael Berks
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - John Gaffney
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Ross A. Little
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Nuria Porta
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Nivetha Sridharan
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Anubhav Datta
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Radiology Department, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Damien J. McHugh
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Christina J. Hague
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Susan Cheung
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Prakash Manoharan
- Radiology Department, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Marcel van Herk
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ananya Choudhury
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Julian C. Matthews
- Division of Psychology, Communication and Human Neuroscience, University of Manchester, Manchester, United Kingdom
| | - Geoff J.M. Parker
- Bioxydyn Ltd, Manchester, United Kingdom
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - David L. Buckley
- Christie Medical Physics and Engineering, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Biomedical Imaging, University of Leeds, Leeds, United Kingdom
| | - Kevin J. Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Andrew McPartlin
- Clinical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Radiation Oncology, Princess Margaret Cancer Center, Toronto, Canada
| | - James P.B. O’Connor
- Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
- Radiology Department, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
7
|
Caldarella C, De Risi M, Massaccesi M, Miccichè F, Bussu F, Galli J, Rufini V, Leccisotti L. Role of 18F-FDG PET/CT in Head and Neck Squamous Cell Carcinoma: Current Evidence and Innovative Applications. Cancers (Basel) 2024; 16:1905. [PMID: 38791983 PMCID: PMC11119768 DOI: 10.3390/cancers16101905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
This article provides an overview of the use of 18F-FDG PET/CT in various clinical scenarios of head-neck squamous cell carcinoma, ranging from initial staging to treatment-response assessment, and post-therapy follow-up, with a focus on the current evidence, debated issues, and innovative applications. Methodological aspects and the most frequent pitfalls in head-neck imaging interpretation are described. In the initial work-up, 18F-FDG PET/CT is recommended in patients with metastatic cervical lymphadenectomy and occult primary tumor; moreover, it is a well-established imaging tool for detecting cervical nodal involvement, distant metastases, and synchronous primary tumors. Various 18F-FDG pre-treatment parameters show prognostic value in terms of disease progression and overall survival. In this scenario, an emerging role is played by radiomics and machine learning. For radiation-treatment planning, 18F-FDG PET/CT provides an accurate delineation of target volumes and treatment adaptation. Due to its high negative predictive value, 18F-FDG PET/CT, performed at least 12 weeks after the completion of chemoradiotherapy, can prevent unnecessary neck dissections. In addition to radiomics and machine learning, emerging applications include PET/MRI, which combines the high soft-tissue contrast of MRI with the metabolic information of PET, and the use of PET radiopharmaceuticals other than 18F-FDG, which can answer specific clinical needs.
Collapse
Affiliation(s)
- Carmelo Caldarella
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
| | - Marina De Risi
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
| | - Mariangela Massaccesi
- Radiation Oncology Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Francesco Miccichè
- Radiation Oncology Unit, Ospedale Isola Tiberina—Gemelli Isola, 00186 Rome, Italy;
| | - Francesco Bussu
- Otorhinolaryngology Operative Unit, Azienda Ospedaliero Universitaria Sassari, 07100 Sassari, Italy;
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Jacopo Galli
- Otorhinolaryngology Unit, Department of Neurosciences, Sensory Organs and Thorax, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Section of Otolaryngology, Department of Head-Neck and Sensory Organs, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Vittoria Rufini
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
- Section of Nuclear Medicine, Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Lucia Leccisotti
- Nuclear Medicine Unit, Department of Radiology and Oncologic Radiotherapy, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (C.C.); (M.D.R.); (L.L.)
- Section of Nuclear Medicine, Department of Radiological Sciences and Hematology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
8
|
Metrard G, Cohen C, Bailly M. Comprehensive literature review of oral and intravenous contrast-enhanced PET/CT: a step forward? Front Med (Lausanne) 2024; 11:1373260. [PMID: 38566921 PMCID: PMC10985176 DOI: 10.3389/fmed.2024.1373260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024] Open
Abstract
The integration of diagnostic CT scans into PET/CT facilitates a comprehensive single examination, presenting potential advantages for patients seeking a thorough one-shot check-up. The introduction of iodinated contrast media during PET scanning raises theoretical concerns about potential interference with uptake quantification, due to the modification of tissue density on CT. Nevertheless, this impact appears generally insignificant for clinical use, compared to the intrinsic variability of standardized uptake values. On the other hand, with the growing indications of PET, especially 18F-FDG PET, contrast enhancement increases the diagnostic performances of the exam, and provides additional information. This improvement in performance achieved through contrast-enhanced PET/CT must be carefully evaluated considering the associated risks and side-effects stemming from the administration of iodinated contrast media. Within this article, we present a comprehensive literature review of contrast enhanced PET/CT, examining the potential impact of iodinated contrast media on quantification, additional side-effects and the pivotal clinically demonstrated benefits of an all-encompassing examination for patients. In conclusion, the clinical benefits of iodinated contrast media are mainly validated by the large diffusion in PET protocols. Contrary to positive oral contrast, which does not appear to offer any major advantage in patient management, intravenous iodine contrast media provides clinical benefits without significant artifact on images or quantification. However, studies on the benefit-risk balance for patients are still lacking.
Collapse
Affiliation(s)
- Gilles Metrard
- Nuclear Medicine Department, Orléans University Hospital, Orléans, France
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Orléans, France
| | - Clara Cohen
- Radiology Department, Orléans University Hospital, Orléans, France
| | - Matthieu Bailly
- Nuclear Medicine Department, Orléans University Hospital, Orléans, France
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Orléans, France
| |
Collapse
|
9
|
Hirata K, Kamagata K, Ueda D, Yanagawa M, Kawamura M, Nakaura T, Ito R, Tatsugami F, Matsui Y, Yamada A, Fushimi Y, Nozaki T, Fujita S, Fujioka T, Tsuboyama T, Fujima N, Naganawa S. From FDG and beyond: the evolving potential of nuclear medicine. Ann Nucl Med 2023; 37:583-595. [PMID: 37749301 DOI: 10.1007/s12149-023-01865-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 09/09/2023] [Indexed: 09/27/2023]
Abstract
The radiopharmaceutical 2-[fluorine-18]fluoro-2-deoxy-D-glucose (FDG) has been dominantly used in positron emission tomography (PET) scans for over 20 years, and due to its vast utility its applications have expanded and are continuing to expand into oncology, neurology, cardiology, and infectious/inflammatory diseases. More recently, the addition of artificial intelligence (AI) has enhanced nuclear medicine diagnosis and imaging with FDG-PET, and new radiopharmaceuticals such as prostate-specific membrane antigen (PSMA) and fibroblast activation protein inhibitor (FAPI) have emerged. Nuclear medicine therapy using agents such as [177Lu]-dotatate surpasses conventional treatments in terms of efficacy and side effects. This article reviews recently established evidence of FDG and non-FDG drugs and anticipates the future trajectory of nuclear medicine.
Collapse
Affiliation(s)
- Kenji Hirata
- Department of Diagnostic Imaging, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| | - Koji Kamagata
- Department of Radiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Daiju Ueda
- Department of Diagnostic and Interventional Radiology, Graduate School of Medicine, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka, 545-8585, Japan
| | - Masahiro Yanagawa
- Department of Radiology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Mariko Kawamura
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Takeshi Nakaura
- Department of Diagnostic Radiology, Kumamoto University Graduate School of Medicine, 1-1-1 Honjo Chuo-ku, Kumamoto, 860-8556, Japan
| | - Rintaro Ito
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| | - Fuminari Tatsugami
- Department of Diagnostic Radiology, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yusuke Matsui
- Department of Radiology, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Akira Yamada
- Department of Radiology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano, 390-2621, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Taiki Nozaki
- Department of Radiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-0016, Japan
| | - Shohei Fujita
- Department of Radiology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Tomoyuki Fujioka
- Department of Diagnostic Radiology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8519, Japan
| | - Takahiro Tsuboyama
- Department of Radiology, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Fujima
- Department of Diagnostic and Interventional Radiology, Hokkaido University Hospital, N15, W5, Kita-ku, Sapporo, 060-8638, Japan
| | - Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan
| |
Collapse
|
10
|
Wei C, Lan X, Qiu M, Cui R, Fu Q, Shinge SAU, Muluh TA, Jiang O. Expanding the role of combined immunochemotherapy and immunoradiotherapy in the management of head and neck cancer (Review). Oncol Lett 2023; 26:372. [PMID: 37965160 PMCID: PMC10641411 DOI: 10.3892/ol.2023.13958] [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: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 11/16/2023] Open
Abstract
Immunotherapy has become one of the most promising approaches in tumor therapy, and there are numerous associated clinical trials in China. As an immunosuppressive tumor, head and neck squamous cell carcinoma (HNSCC) carries a high mutation burden, making immune checkpoint inhibitors promising candidates in this field due to their unique mechanism of action. The present review outlines a comprehensive multidisciplinary cancer treatment approach and elaborates on how combining immunochemotherapy and immunoradiotherapy guidelines could enhance clinical efficacy in patients with HNSCC. Furthermore, the present review explores the immunology of HNSCC, current immunotherapeutic strategies to enhance antitumor activity, ongoing clinical trials and the future direction of the current immune landscape in HNSCC. Advanced-stage HNSCC presents with a poor prognosis, low survival rates and minimal improvement in patient survival trends over time. Understanding the potential of immunotherapy and ways to combine it with surgery, chemotherapy and radiotherapy confers good prospects for the management of human papillomavirus (HPV)-positive HNSCC, as well as other HPV-positive malignancies. Understanding the immune system and its effect on HNSCC progression and metastasis will help to uncover novel biomarkers for the selection of patients and to enhance the efficacy of treatments. Further research on why current immune checkpoint inhibitors and targeted drugs are only effective for some patients in the clinic is needed; therefore, further research is required to improve the overall survival of affected patients.
Collapse
Affiliation(s)
- Chun Wei
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Xiaojun Lan
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Maona Qiu
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| | - Qiuxia Fu
- Department of General Medicine, The People's Hospital of Luzhou City, Luzhou, Sichuan 646000, P.R. China
| | - Shafiu A. Umar Shinge
- Department of Cardiothoracic Surgery, Sun Yat Sen Memorial Hospital, Sun Yat Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong 518060, P.R. China
| | - Ou Jiang
- Department of Oncology, The Second People's Hospital of Neijiang City, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
11
|
Kjems J, Elisabet Håkansson K, Andrup Kristensen C, Grau Eriksen J, Horsholt Kristensen M, Ivalu Sander Holm A, Overgaard J, Rønn Hansen C, Zukauskaite R, Johansen J, Richter Vogelius I, Friborg J. The influence of tumor volume on the risk of distant metastases in head and neck squamous cell carcinomas. Radiother Oncol 2023; 186:109771. [PMID: 37385382 DOI: 10.1016/j.radonc.2023.109771] [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: 03/16/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/01/2023]
Abstract
BACKGROUND AND PURPOSE Distant metastases (DM) in head and neck squamous cell carcinomas (HNSCC) are in most circumstances non-curable. The TNM staging system is insufficient to predict the risk of DM. This study investigates if the DM risk can be predicted using a multivariate model including pre-treatment total tumor volume for both p16-positive oropharyngeal squamous cell carcinoma (OPSCC) and all other sites (other HNSCC). MATERIALS AND METHODS The study includes patients with localized pharyngeal and laryngeal squamous cell carcinomas treated with primary radiotherapy from 2008-2017 from three head and neck cancer centers. Patients were identified in the Danish Head and Neck Cancer (DAHANCA) database. Total (nodal and primary) tumor volume (Gross Tumor Volume, GTV) was extracted from local treatment planning systems. The GTV was grouped by volume (cm3) in four intervals and included in a multivariate Cox proportional hazard regression controlled for pre-selected clinical values incl. stage. RESULTS The study includes 2,865 patients, of which 321 (11 %) had DM post-treatment. The risk of DM was assessed in a multivariate model based on 2,751 patients (p16-positive OPSCC: 1,032; and other HNSCC: 1,719). There was a significant association between GTV and the risk of DM, and in tumor volumes ≥ 50 cm3 hazard ratios of 7.6 (2.5-23.4) for p16-positive OPSCC and 4.1 (2.3-7.2) in other HNSCC were observed. CONCLUSION Tumor volume is an independent risk factor for DM. The addition of total tumor volume to a predictive model is important to identify subgroups of HNSCC patients at high risk of DM.
Collapse
Affiliation(s)
- Julie Kjems
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen 2100, Denmark.
| | | | - Claus Andrup Kristensen
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen 2100, Denmark.
| | - Jesper Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus 8200, Denmark.
| | | | | | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus 8200, Denmark.
| | - Christian Rønn Hansen
- Department of Oncology, Odense University Hospital, Odense 5000, Denmark; Laboratory of Radiation Physics, Odense University Hospital, Odense 5000, Denmark; Department of Clinical Research, University of Southern Denmark, Odense 5000, Denmark.
| | - Ruta Zukauskaite
- Department of Oncology, Odense University Hospital, Odense 5000, Denmark.
| | - Jørgen Johansen
- Department of Oncology, Odense University Hospital, Odense 5000, Denmark.
| | - Ivan Richter Vogelius
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen 2100, Denmark.
| | - Jeppe Friborg
- Department of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen 2100, Denmark.
| |
Collapse
|
12
|
Boeke S, Winter RM, Leibfarth S, Krueger MA, Bowden G, Cotton J, Pichler BJ, Zips D, Thorwarth D. Machine learning identifies multi-parametric functional PET/MR imaging cluster to predict radiation resistance in preclinical head and neck cancer models. Eur J Nucl Med Mol Imaging 2023; 50:3084-3096. [PMID: 37148296 PMCID: PMC10382355 DOI: 10.1007/s00259-023-06254-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 04/25/2023] [Indexed: 05/08/2023]
Abstract
PURPOSE Tumor hypoxia and other microenvironmental factors are key determinants of treatment resistance. Hypoxia positron emission tomography (PET) and functional magnetic resonance imaging (MRI) are established prognostic imaging modalities to identify radiation resistance in head-and-neck cancer (HNC). The aim of this preclinical study was to develop a multi-parametric imaging parameter specifically for focal radiotherapy (RT) dose escalation using HNC xenografts of different radiation sensitivities. METHODS A total of eight human HNC xenograft models were implanted into 68 immunodeficient mice. Combined PET/MRI using dynamic [18F]-fluoromisonidazole (FMISO) hypoxia PET, diffusion-weighted (DW), and dynamic contrast-enhanced MRI was carried out before and after fractionated RT (10 × 2 Gy). Imaging data were analyzed on voxel-basis using principal component (PC) analysis for dynamic data and apparent diffusion coefficients (ADCs) for DW-MRI. A data- and hypothesis-driven machine learning model was trained to identify clusters of high-risk subvolumes (HRSs) from multi-dimensional (1-5D) pre-clinical imaging data before and after RT. The stratification potential of each 1D to 5D model with respect to radiation sensitivity was evaluated using Cohen's d-score and compared to classical features such as mean/peak/maximum standardized uptake values (SUVmean/peak/max) and tumor-to-muscle-ratios (TMRpeak/max) as well as minimum/valley/maximum/mean ADC. RESULTS Complete 5D imaging data were available for 42 animals. The final preclinical model for HRS identification at baseline yielding the highest stratification potential was defined in 3D imaging space based on ADC and two FMISO PCs ([Formula: see text]). In 1D imaging space, only clusters of ADC revealed significant stratification potential ([Formula: see text]). Among all classical features, only ADCvalley showed significant correlation to radiation resistance ([Formula: see text]). After 2 weeks of RT, FMISO_c1 showed significant correlation to radiation resistance ([Formula: see text]). CONCLUSION A quantitative imaging metric was described in a preclinical study indicating that radiation-resistant subvolumes in HNC may be detected by clusters of ADC and FMISO using combined PET/MRI which are potential targets for future functional image-guided RT dose-painting approaches and require clinical validation.
Collapse
Affiliation(s)
- Simon Boeke
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - René M Winter
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Sara Leibfarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany
| | - Marcel A Krueger
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Gregory Bowden
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Jonathan Cotton
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Hoppe-Seyler-Str. 3, 72076, Tübingen, Germany.
| |
Collapse
|
13
|
Perez RC, Kim D, Maxwell AWP, Camacho JC. Functional Imaging of Hypoxia: PET and MRI. Cancers (Basel) 2023; 15:3336. [PMID: 37444446 DOI: 10.3390/cancers15133336] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/22/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Molecular and functional imaging have critical roles in cancer care. Existing evidence suggests that noninvasive detection of hypoxia within a particular type of cancer can provide new information regarding the relationship between hypoxia, cancer aggressiveness and altered therapeutic responses. Following the identification of hypoxia inducible factor (HIF), significant progress in understanding the regulation of hypoxia-induced genes has been made. These advances have provided the ability to therapeutically target HIF and tumor-associated hypoxia. Therefore, by utilizing the molecular basis of hypoxia, hypoxia-based theranostic strategies are in the process of being developed which will further personalize care for cancer patients. The aim of this review is to provide an overview of the significance of tumor hypoxia and its relevance in cancer management as well as to lay out the role of imaging in detecting hypoxia within the context of cancer.
Collapse
Affiliation(s)
- Ryan C Perez
- Florida State University College of Medicine, Tallahassee, FL 32306, USA
| | - DaeHee Kim
- Department of Diagnostic Imaging, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Aaron W P Maxwell
- Department of Diagnostic Imaging, The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Juan C Camacho
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306, USA
| |
Collapse
|
14
|
Lin Fracp P, Holloway L, Min Franzcr M, Lee Franzcr M, Fowler Franzcr A. Prognostic and predictive values of baseline and mid-treatment FDG-PET in oropharyngeal carcinoma treated with primary definitive (chemo)radiation and impact of HPV status: review of current literature and emerging roles. Radiother Oncol 2023; 184:109686. [PMID: 37142128 DOI: 10.1016/j.radonc.2023.109686] [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: 07/29/2022] [Revised: 04/16/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND AND PURPOSE This study provides a review of the literature assessing whether semiquantitative PET parameters acquired at baseline and/or during definitive (chemo)radiotherapy ("prePET" and "iPET") can predict survival outcomes in patients with oropharyngeal squamous cell carcinoma (OPC), and the impact of human papilloma virus (HPV) status. MATERIAL AND METHODS A literature search was carried out using PubMed and Embase between 2001 to 2021 in accordance with PRISMA. RESULTS The analysis included 22 FDG-PET/CT studies1-22, 19 pre-PET and 3 both pre-PET and iPET14,18,20,. The analysis involved 2646 patients, of which 1483 are HPV-positive (17 studies: 10 mixed and 7 HPV-positive only), 589 are HPV-negative, and 574 have unknown HPV status. Eighteen studies found significant correlations of survival outcomes with pre-PET parameters, most commonly primary or "Total" (combined primary and nodal) metabolic tumour volume and/or total lesional glycolysis. Two studies could not establish significant correlations and both employed SUVmax only. Two studies also could not establish significant correlations when taking into account of the HPV-positive population only. Because of the heterogeneity and lack of standardized methodology, no conclusions on optimal cut-off values can be drawn. Ten studies specifically evaluated HPV-positive patients: five showed positive correlation of pre-PET parameters and survival outcomes, but four of these studies did not include advanced T or N staging in multivariate analysis1,6,15,22, and two studies only showed positive correlations after excluding high risk patients with smoking history7 or adverse CT features22. Two studies found that prePET parameters predicted treatment outcomes only in HPV-negative but not HPV-positive patients10,16. Two studies found that iPET parameters could predict outcomes in HPV-positive patients but not prePET parameters14,18. CONCLUSION The current literature supports high pre-treatment metabolic burden prior to definitive (chemo)radiotherapy can predict poor treatment outcomes for HPV-negative OPC patients. Evidence is conflicting and currently does not support correlation in HPV-positive patients.
Collapse
Affiliation(s)
- Peter Lin Fracp
- Department of Nuclear Medicine and PET, Liverpool Hospital, Liverpool, NSW, Australia; South Western Sydney Clinical School, University of New South Wales, NSW, Australia; School of Medicine, Western Sydney University, NSW, Australia.
| | - Lois Holloway
- South Western Sydney Clinical School, University of New South Wales, NSW, Australia; School of Medicine, Western Sydney University, NSW, Australia; Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW, Australia; Ingham Institute of Applied Medical Research, Liverpool, NSW, Australia
| | - Myo Min Franzcr
- Department of Radiation Oncology, Sunshine Coast University Hospital, Queensland, Australia; Faculty of Science, Health, Education and Engineering, University of Sunshine Coast, Queensland, Australia
| | - Mark Lee Franzcr
- South Western Sydney Clinical School, University of New South Wales, NSW, Australia; Cancer Therapy Centre, Liverpool Hospital, Liverpool, NSW, Australia
| | | |
Collapse
|
15
|
Thorwarth D. Clinical use of positron emission tomography for radiotherapy planning - Medical physics considerations. Z Med Phys 2023; 33:13-21. [PMID: 36272949 PMCID: PMC10068574 DOI: 10.1016/j.zemedi.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 08/17/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022]
Abstract
PET/CT imaging plays an increasing role in radiotherapy treatment planning. The aim of this article was to identify the major use cases and technical as well as medical physics challenges during integration of these data into treatment planning. Dedicated aspects, such as (i) PET/CT-based radiotherapy simulation, (ii) PET-based target volume delineation, (iii) functional avoidance to optimized organ-at-risk sparing and (iv) functionally adapted individualized radiotherapy are discussed in this article. Furthermore, medical physics aspects to be taken into account are summarized and presented in form of check-lists.
Collapse
Affiliation(s)
- Daniela Thorwarth
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen; and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
16
|
Hildingsson S, Gebre-Medhin M, Zschaeck S, Adrian G. Hypoxia in relationship to tumor volume using hypoxia PET-imaging in head & neck cancer - A scoping review. Clin Transl Radiat Oncol 2022; 36:40-46. [PMID: 35769424 PMCID: PMC9234341 DOI: 10.1016/j.ctro.2022.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023] Open
Abstract
Background Hypoxia and large tumor volumes are negative prognostic factors for patients with head and neck squamous cell carcinoma (HNSCC) treated with radiation therapy (RT). PET-scanning with specific hypoxia-tracers (hypoxia-PET) can be used to non-invasively assess hypoxic tumor volume. Primary tumor volume is readily available for patients undergoing RT. However, the relationship between hypoxic volume and primary tumor volume is yet an open question. The current study investigates the hypotheses that larger tumors contain both a larger hypoxic volume and a higher hypoxic fraction. Methods PubMed and Embase were systematically searched to identify articles fulfilling the predefined criteria. Individual tumor data (primary tumor volume and hypoxic volume/fraction) was extracted. Relationship between hypoxic volume and primary tumor volume was investigated by linear regression. The correlation between hypoxic fraction and log2(primary tumor volume) was determined for each cohort and in a pooled analysis individual regression slopes and coefficients of determination (R2) were weighted according to cohort size. Results 21 relevant articles were identified and individual data from 367 patients was extracted, out of which 323 patients from 17 studies had quantifiable volumes of interest. A correlation between primary tumor volume and PET-determined hypoxic volume was found (P <.001, R2 = 0.46). Larger tumors had a significantly higher fraction of hypoxia compared with smaller tumors (P<.01). The weighted analysis of all studies revealed that for each doubling of the tumor volume, the hypoxic fraction increased by four percentage points. Conclusion This study shows correlations between primary tumor volume and hypoxic volume as well as primary tumor volume and the hypoxic fraction in patients with HNSCC. The findings suggest that not only do large tumors contain more cancer cells, they also have a higher proportion of potentially radioresistant hypoxic cells. This knowledge can be important when individualizing RT.
Collapse
Affiliation(s)
- Sofia Hildingsson
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Gebre-Medhin
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gabriel Adrian
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
17
|
Lefèvre AC, Alsner J, Sørensen BS, Tramm T, Toustrup K, Overgaard J, Spindler KLG. Hypoxia and local tumour control in squamous cell carcinoma of the anus - a hypothesis-generating study. Acta Oncol 2022; 61:1132-1135. [PMID: 35730972 DOI: 10.1080/0284186x.2022.2089591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Anna Cecilie Lefèvre
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Brita Singers Sørensen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - Kasper Toustrup
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Karen-Lise Garm Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
18
|
Alsner J, Overgaard J, Tramm T, Lindegaard JC. Hypoxic gene expression is a prognostic factor for disease free survival in a cohort of locally advanced squamous cell cancer of the uterine cervix. Acta Oncol 2022; 61:172-178. [PMID: 34586924 DOI: 10.1080/0284186x.2021.1979249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Tumour hypoxia in locally advanced squamous cervical cancer (LACC) has been shown to be of substantial prognostic importance. The aims of the present study were therefore to investigate if hypoxia could be identified by a newly validated hypoxic gene expression classifier and used as a prognostic factor for disease free survival (DFS). MATERIAL AND METHODS Paraffin embedded biopsies were obtained from 190 patients with LACC with squamous cell carcinoma treated 2005-2016 with chemo-radiation and image guided adaptive brachytherapy. Analysis of hypoxia was successful in 183 patients (96%). Hypoxic classification of tumours into 'more' or 'less' hypoxic was based on 15 genes using the same method as in a prospective head and neck cancer trial (NCT02661152). HPV was genotyped using INNO-LiPA. Local tumour invasion was evaluated by the T-score. Primary endpoint was DFS analysed by Kaplan-Meier and Cox regression. Events were death of any cause, persistent disease, or recurrence. RESULTS The FIGO2009 stage distribution was IB-IIA 9%, IIB 64%, and III-IVA 27%, and mean T-score was 7.2. Pathological nodes were present in 53%. Median observation time was 5.2 years. Local control rate at 5 years was 96%, and pelvic (loco-regional) control 91%. Overall, 36% of the tumours were classified as 'more' hypoxic. The frequency of 'more' hypoxic tumours increased with local tumour intrusion (30% for T-score 0-9 vs. 55% for T-score ≥10, p = 0.004). Hypoxia was associated with decreased DFS in univariate, HR 1.71 (1.04-2.82), and multivariate analysis, HR 1.75 (1.04-2.92), and the effect was particularly observed among tumours with a T-score ≥10. HPV 16/18 was not associated with improved DFS in neither in univariate nor in multivariate analysis. CONCLUSION Hypoxic gene expression is a prominent prognostic factor for DFS in LACC with SCC histology and should be considered for treatment stratification in clinical trials.
Collapse
Affiliation(s)
- Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Trine Tramm
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
19
|
den bossche VV, Zaryouh H, Vara-Messler M, Vignau J, Machiels JP, Wouters A, Schmitz S, Corbet C. Microenvironment-driven intratumoral heterogeneity in head and neck cancers: clinical challenges and opportunities for precision medicine. Drug Resist Updat 2022; 60:100806. [DOI: 10.1016/j.drup.2022.100806] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/20/2022] [Accepted: 01/21/2022] [Indexed: 02/06/2023]
|
20
|
Therapeutic targeting of the hypoxic tumour microenvironment. Nat Rev Clin Oncol 2021; 18:751-772. [PMID: 34326502 DOI: 10.1038/s41571-021-00539-4] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Hypoxia is prevalent in human tumours and contributes to microenvironments that shape cancer evolution and adversely affect therapeutic outcomes. Historically, two different tumour microenvironment (TME) research communities have been discernible. One has focused on physicochemical gradients of oxygen, pH and nutrients in the tumour interstitium, motivated in part by the barrier that hypoxia poses to effective radiotherapy. The other has focused on cellular interactions involving tumour and non-tumour cells within the TME. Over the past decade, strong links have been established between these two themes, providing new insights into fundamental aspects of tumour biology and presenting new strategies for addressing the effects of hypoxia and other microenvironmental features that arise from the inefficient microvascular system in solid tumours. This Review provides a perspective on advances at the interface between these two aspects of the TME, with a focus on translational therapeutic opportunities relating to the elimination and/or exploitation of tumour hypoxia.
Collapse
|
21
|
Lassen P, Huang SH, Su J, Waldron J, Andersen M, Primdahl H, Johansen J, Kristensen CA, Andersen E, Eriksen JG, Hansen CR, Alsner J, Lilja-Fisher J, Bratman SV, Ringash J, Kim J, Hope A, Spreafico A, de Almeida J, Xu W, O'Sullivan B, Overgaard J. Treatment outcomes and survival following definitive (chemo)radiotherapy in HPV-positive oropharynx cancer: Large-scale comparison of DAHANCA vs PMH cohorts. Int J Cancer 2021; 150:1329-1340. [PMID: 34792199 DOI: 10.1002/ijc.33876] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 10/17/2021] [Accepted: 11/02/2021] [Indexed: 11/05/2022]
Abstract
We compare outcomes in two large-scale contemporaneously treated HPV-positive (HPV+) oropharynx cancer (OPC) cohorts treated with definitive radiotherapy/chemoradiotherapy (RT/CRT). p16-confirmed HPV+ OPC treated between 2007 and 2015 at PMH and DAHANCA were identified. Locoregional failure (LRF), distant metastasis (DM), and overall survival (OS) were compared. Multivariable analysis (MVA) calculated adjusted-hazard-ratio (aHR) with 95% confidence interval (95% CI), adjusting for cohort, age, gender, performance status, smoking pack-years, T-category and N-category and chemotherapy. Compared to PMH (n = 701), DAHANCA (n = 1174) contained lower TNM-8T-categories (T1-T2: 77% vs 56%), N-categories (N0-N1: 77% vs 67%) and stages (stage I: 63% vs 44% (all P < .001). PMH used standard-fractionation CRT in 69% (481) while 31% (220) received hypofractionated or moderately accelerated RT-alone. All DAHANCA patients were treated with moderately accelerated RT; 96% (1129) received nimorazole (NIM) and 73% (856) concurrent weekly cisplatin. DAHANCA had shorter overall-treatment-time (P < .001), lower gross tumor (66-68 vs 70 Gy) and elective neck (50 vs 56 Gy) doses. Median follow-up was 4.8 years. DAHANCA had higher 5-year LRF (13% vs 7%, aHR = 0.47 [0.34-0.67]), comparable DM (7% vs 12%, aHR = 1.32 [0.95-1.82]), but better OS (85% vs 80%, aHR = 1.30 [1.01-1.68]). CRT patients had a lower risk of LRF (aHR 0.56 [0.39-0.82]), DM (aHR 0.70 [0.50-1.00]) and death (aHR 0.39 [0.29-0.52]) vs RT-alone. We observed exemplary outcomes for two large-scale trans-Atlantic HPV+ OPC cohorts treated in a similar manner. Concurrent chemotherapy was a strong, independent prognostic factor for all endpoints. Our findings underscore the need for a very careful approach to de-intensification of treatment for this disease.
Collapse
Affiliation(s)
- Pernille Lassen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Shao Hui Huang
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Jie Su
- Department of Biostatistics, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - John Waldron
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Maria Andersen
- Department of Oncology, Aalborg Univeristy Hospital, Aalborg, Denmark
| | - Hanne Primdahl
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jørgen Johansen
- Department of Oncology, Odense Universitetshospital, Odense, Denmark
| | | | - Elo Andersen
- Department of Oncology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Grau Eriksen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Jan Alsner
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob Lilja-Fisher
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Scott V Bratman
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Jolie Ringash
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - John Kim
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Andrew Hope
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Division of Medical Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - John de Almeida
- Department of Otolaryngology - Head & Neck Surgery, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Wei Xu
- Department of Biostatistics, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Brian O'Sullivan
- Department of Radiation Oncology, Princess Margaret Hospital Cancer Centre/University of Toronto, Toronto, Ontario, Canada
| | - Jens Overgaard
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
22
|
Florea A, Mottaghy FM, Bauwens M. Molecular Imaging of Angiogenesis in Oncology: Current Preclinical and Clinical Status. Int J Mol Sci 2021; 22:5544. [PMID: 34073992 PMCID: PMC8197399 DOI: 10.3390/ijms22115544] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/11/2022] Open
Abstract
Angiogenesis is an active process, regulating new vessel growth, and is crucial for the survival and growth of tumours next to other complex factors in the tumour microenvironment. We present possible molecular imaging approaches for tumour vascularisation and vitality, focusing on radiopharmaceuticals (tracers). Molecular imaging in general has become an integrated part of cancer therapy, by bringing relevant insights on tumour angiogenic status. After a structured PubMed search, the resulting publication list was screened for oncology related publications in animals and humans, disregarding any cardiovascular findings. The tracers identified can be subdivided into direct targeting of angiogenesis (i.e., vascular endothelial growth factor, laminin, and fibronectin) and indirect targeting (i.e., glucose metabolism, hypoxia, and matrix metallo-proteases, PSMA). Presenting pre-clinical and clinical data of most tracers proposed in the literature, the indirect targeting agents are not 1:1 correlated with angiogenesis factors but do have a strong prognostic power in a clinical setting, while direct targeting agents show most potential and specificity for assessing tumour vascularisation and vitality. Within the direct agents, the combination of multiple targeting tracers into one agent (multimers) seems most promising. This review demonstrates the present clinical applicability of indirect agents, but also the need for more extensive research in the field of direct targeting of angiogenesis in oncology. Although there is currently no direct tracer that can be singled out, the RGD tracer family seems to show the highest potential therefore we expect one of them to enter the clinical routine.
Collapse
Affiliation(s)
- Alexandru Florea
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, 6229HX Maastricht, The Netherlands
| | - Felix M. Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht University, 6229HX Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229HX Maastricht, The Netherlands
| | - Matthias Bauwens
- Department of Nuclear Medicine, University Hospital RWTH Aachen, 52074 Aachen, Germany; (A.F.); (M.B.)
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, 6229HX Maastricht, The Netherlands
- School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6229HX Maastricht, The Netherlands
| |
Collapse
|