1
|
Kafkaletos A, Mix M, Sachpazidis I, Carles M, Rühle A, Ruf J, Grosu AL, Nicolay NH, Baltas D. The significance of partial volume effect on the estimation of hypoxic tumour volume with [ 18F]FMISO PET/CT. EJNMMI Phys 2024; 11:43. [PMID: 38722446 PMCID: PMC11082115 DOI: 10.1186/s40658-024-00643-1] [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: 01/16/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The purpose of this study was to evaluate how a retrospective correction of the partial volume effect (PVE) in [18F]fluoromisonidazole (FMISO) PET imaging, affects the hypoxia discoverability within a gross tumour volume (GTV). This method is based on recovery coefficients (RC) and is tailored for low-contrast tracers such as FMISO. The first stage was the generation of the scanner's RC curves, using spheres with diameters from 10 to 37 mm, and the same homogeneous activity concentration, positioned in lower activity concentration background. Six sphere-to-background contrast ratios were used, from 10.0:1, down to 2.0:1, in order to investigate the dependence of RC on both the volume and the contrast ratio. The second stage was to validate the recovery-coefficient correction method in a more complex environment of non-spherical lesions of different volumes and inhomogeneous activity concentration. Finally, we applied the correction method to a clinical dataset derived from a prospective imaging trial (DRKS00003830): forty nine head and neck squamous cell carcinoma (HNSCC) cases who had undergone FMISO PET/CT scanning for the quantification of tumour hypoxia before (W0), 2 weeks (W2) and 5 weeks (W5) after the beginning of radiotherapy. Here, PVE was found to cause an underestimation of the activity in small volumes with high FMISO signal. RESULTS The application of the proposed correction method resulted in a statistically significant increase of both the hypoxic subvolume (171% at W0, 691% at W2 and 4.60 × 103% at W5 with p < 0.001) and the FMISO standardised uptake value (SUV) (27% at W0, 21% at W2 and by 25% at W5 with p < 0.001) within the primary GTV. CONCLUSIONS The proposed PVE-correction method resulted in a statistically significant increase of the hypoxic fraction (HF) with p < 0.001 and demonstrated results in better agreement with published HF data for HNSCC. To summarise, the proposed RC-based correction method can be a useful tool for a retrospective compensation against PVE.
Collapse
Affiliation(s)
- Athanasios Kafkaletos
- Division of Medical Physics, Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany.
| | - Michael Mix
- Department of Nuclear Medicine, Medical Centre - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
| | - Ilias Sachpazidis
- Division of Medical Physics, Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
| | - Montserrat Carles
- Biomedical Imaging Research Group (GIBI230-PREBI) and Imaging La Fe Node at Distributed Network for Biomedical Imaging (ReDIB) Unique Scientific and Technical Infrastructures (ICTS), La Fe Health Research Institute, Valencia, Spain
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
- Department of Radiation Oncology, University of Leipzig Medical Centre, Leipzig, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Centre - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
- Department of Radiation Oncology, University of Leipzig Medical Centre, Leipzig, Germany
| | - Dimos Baltas
- Division of Medical Physics, Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, German Cancer Consortium (DKTK), partner site DKTK-Freiburg, University of Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Sambasivan K, Barrington SF, Connor SE, Witney TH, Blower PJ, Urbano TG. Is there a role for [ 18F]-FMISO PET to guide dose adaptive radiotherapy in head and neck cancer? A review of the literature. Clin Transl Imaging 2024; 12:137-155. [PMID: 39286295 PMCID: PMC7616449 DOI: 10.1007/s40336-023-00607-y] [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: 10/03/2023] [Accepted: 12/12/2023] [Indexed: 09/19/2024]
Abstract
Purpose Hypoxia is a major cause of radioresistance in head and neck cancer (HNC), resulting in treatment failure and disease recurrence. 18F-fluoromisonidazole [18F]FMISO PET has been proposed as a means of localising intratumoural hypoxia in HNC so that radiotherapy can be specifically escalated in hypoxic regions. This concept may not be deliverable in routine clinical practice, however, given that [18F]FMISO PET is costly, time consuming and difficult to access. The aim of this review was to summarise clinical studies involving [18F]FMISO PET to ascertain whether it can be used to guide radiotherapy treatment in HNC. Methods A comprehensive literature search was conducted on PubMed and Web of Science databases. Studies investigating [18F]FMISO PET in newly diagnosed HNC patients were considered eligible for review. Results We found the following important results from our literature review: 1)Studies have focussed on comparing [18F]FMISO PET to other hypoxia biomarkers, but currently there is no evidence of a strong correlation between [18F]FMISO and these biomarkers.2)The results of [18F]FMISO PET imaging are not necessarily repeatable, and the location of uptake may vary during treatment.3)Tumour recurrences do not always occur within the pretreatment hypoxic volume on [18F]FMISO PET.4)Dose modification studies using [18F]FMISO PET are in a pilot phase and so far, none have demonstrated the efficacy of radiotherapy dose painting according to [18F]FMISO uptake on PET. Conclusions Our results suggest it is unlikely [18F]FMISO PET will be suitable for radiotherapy dose adaptation in HNC in a routine clinical setting. Part of the problem is that hypoxia is a dynamic phenomenon, and thus difficult to delineate on a single scan. Currently, it is anticipated that [18F]FMISO PET will remain useful within the research setting only.
Collapse
Affiliation(s)
- Khrishanthne Sambasivan
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - Sally F Barrington
- King's College London and Guy's and St Thomas' PET Centre; School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, London, UK
| | - Steve Ej Connor
- Department of Neuroradiology, King's College Hospital NHS Foundation Trust, London, UK Department of Radiology, Guy's and St Thomas' NHS Foundation Trust, London, UK; School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, King's College London, London, UK
| | - Timothy H Witney
- King's College London, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, London, United Kingdom
| | - Philip J Blower
- King's College London, School of Biomedical Engineering and Imaging Sciences, St Thomas' Hospital, London, United Kingdom
| | - Teresa Guerrero Urbano
- Department of Clinical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK; Faculty of Dentistry, Oral & Craniofacial Sciences and School of Cancer & Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
3
|
Gertsenshteyn I, Epel B, Ahluwalia A, Kim H, Fan X, Barth E, Zamora M, Markiewicz E, Tsai HM, Sundramoorthy S, Leoni L, Lukens J, Bhuiyan M, Freifelder R, Kucharski A, Giurcanu M, Roman BB, Karczmar G, Kao CM, Halpern H, Chen CT. The optimal 18F-fluoromisonidazole PET threshold to define tumor hypoxia in preclinical squamous cell carcinomas using pO 2 electron paramagnetic resonance imaging as reference truth. Eur J Nucl Med Mol Imaging 2022; 49:4014-4024. [PMID: 35792927 PMCID: PMC9529789 DOI: 10.1007/s00259-022-05889-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/19/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE To identify the optimal threshold in 18F-fluoromisonidazole (FMISO) PET images to accurately locate tumor hypoxia by using electron paramagnetic resonance imaging (pO2 EPRI) as ground truth for hypoxia, defined by pO2 [Formula: see text] 10 mmHg. METHODS Tumor hypoxia images in mouse models of SCCVII squamous cell carcinoma (n = 16) were acquired in a hybrid PET/EPRI imaging system 2 h post-injection of FMISO. T2-weighted MRI was used to delineate tumor and muscle tissue. Dynamic contrast enhanced (DCE) MRI parametric images of Ktrans and ve were generated to model tumor vascular properties. Images from PET/EPR/MRI were co-registered and resampled to isotropic 0.5 mm voxel resolution for analysis. PET images were converted to standardized uptake value (SUV) and tumor-to-muscle ratio (TMR) units. FMISO uptake thresholds were evaluated using receiver operating characteristic (ROC) curve analysis to find the optimal FMISO threshold and unit with maximum overall hypoxia similarity (OHS) with pO2 EPRI, where OHS = 1 shows perfect overlap and OHS = 0 shows no overlap. The means of dice similarity coefficient, normalized Hausdorff distance, and accuracy were used to define the OHS. Monotonic relationships between EPRI/PET/DCE-MRI were evaluated with the Spearman correlation coefficient ([Formula: see text]) to quantify association of vasculature on hypoxia imaged with both FMISO PET and pO2 EPRI. RESULTS FMISO PET thresholds to define hypoxia with maximum OHS (both OHS = 0.728 [Formula: see text] 0.2) were SUV [Formula: see text] 1.4 [Formula: see text] SUVmean and SUV [Formula: see text] 0.6 [Formula: see text] SUVmax. Weak-to-moderate correlations (|[Formula: see text]|< 0.70) were observed between PET/EPRI hypoxia images with vascular permeability (Ktrans) or fractional extracellular-extravascular space (ve) from DCE-MRI. CONCLUSION This is the first in vivo comparison of FMISO uptake with pO2 EPRI to identify the optimal FMISO threshold to define tumor hypoxia, which may successfully direct hypoxic tumor boosts in patients, thereby enhancing tumor control.
Collapse
Affiliation(s)
- Inna Gertsenshteyn
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | - Boris Epel
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | | | - Heejong Kim
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Xiaobing Fan
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Eugene Barth
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | - Marta Zamora
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Erica Markiewicz
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Hsiu-Ming Tsai
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Subramanian Sundramoorthy
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | - Lara Leoni
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - John Lukens
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | - Mohammed Bhuiyan
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | | | - Anna Kucharski
- Department of Radiology, The University of Chicago, Chicago, IL, USA
| | - Mihai Giurcanu
- Department of Public Health Sciences, The University of Chicago, Chicago, IL, USA
| | - Brian B Roman
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Gregory Karczmar
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Chien-Min Kao
- Department of Radiology, The University of Chicago, Chicago, IL, USA
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA
| | - Howard Halpern
- Department of Radiation and Cellular Oncology, The University of Chicago, Chicago, IL, USA
- Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, IL, USA
| | - Chin-Tu Chen
- Department of Radiology, The University of Chicago, Chicago, IL, USA.
- Integrated Small Animal Imaging Research Resource, OSRF, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
4
|
Hildingsson S, Gebre-Medhin M, Zschaeck S, Adrian G. Hypoxia in relationship to tumor volume using hypoxia PET-imaging in head & neck cancer - A scoping review. Clin Transl Radiat Oncol 2022; 36:40-46. [PMID: 35769424 PMCID: PMC9234341 DOI: 10.1016/j.ctro.2022.06.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 01/19/2023] Open
Abstract
Primary tumor volume and hypoxic volume has previously not been convincingly related. 367 patients with head and neck squamous cell carcinoma from 21 different studies using hypoxia-PET The hypoxic volume increased significantly with primary tumor volume. In larger tumor the hypoxic fraction was significantly higher than in smaller tumors.
Background Hypoxia and large tumor volumes are negative prognostic factors for patients with head and neck squamous cell carcinoma (HNSCC) treated with radiation therapy (RT). PET-scanning with specific hypoxia-tracers (hypoxia-PET) can be used to non-invasively assess hypoxic tumor volume. Primary tumor volume is readily available for patients undergoing RT. However, the relationship between hypoxic volume and primary tumor volume is yet an open question. The current study investigates the hypotheses that larger tumors contain both a larger hypoxic volume and a higher hypoxic fraction. Methods PubMed and Embase were systematically searched to identify articles fulfilling the predefined criteria. Individual tumor data (primary tumor volume and hypoxic volume/fraction) was extracted. Relationship between hypoxic volume and primary tumor volume was investigated by linear regression. The correlation between hypoxic fraction and log2(primary tumor volume) was determined for each cohort and in a pooled analysis individual regression slopes and coefficients of determination (R2) were weighted according to cohort size. Results 21 relevant articles were identified and individual data from 367 patients was extracted, out of which 323 patients from 17 studies had quantifiable volumes of interest. A correlation between primary tumor volume and PET-determined hypoxic volume was found (P <.001, R2 = 0.46). Larger tumors had a significantly higher fraction of hypoxia compared with smaller tumors (P<.01). The weighted analysis of all studies revealed that for each doubling of the tumor volume, the hypoxic fraction increased by four percentage points. Conclusion This study shows correlations between primary tumor volume and hypoxic volume as well as primary tumor volume and the hypoxic fraction in patients with HNSCC. The findings suggest that not only do large tumors contain more cancer cells, they also have a higher proportion of potentially radioresistant hypoxic cells. This knowledge can be important when individualizing RT.
Collapse
Affiliation(s)
- Sofia Hildingsson
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden
| | - Maria Gebre-Medhin
- Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Gabriel Adrian
- Division of Oncology and Pathology, Clinical Sciences, Lund University, Lund, Sweden.,Department of Hematology, Oncology and Radiation Physics, Skåne University Hospital, Lund University, Lund, Sweden
| |
Collapse
|
5
|
Gallez B. The Role of Imaging Biomarkers to Guide Pharmacological Interventions Targeting Tumor Hypoxia. Front Pharmacol 2022; 13:853568. [PMID: 35910347 PMCID: PMC9335493 DOI: 10.3389/fphar.2022.853568] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is a common feature of solid tumors that contributes to angiogenesis, invasiveness, metastasis, altered metabolism and genomic instability. As hypoxia is a major actor in tumor progression and resistance to radiotherapy, chemotherapy and immunotherapy, multiple approaches have emerged to target tumor hypoxia. It includes among others pharmacological interventions designed to alleviate tumor hypoxia at the time of radiation therapy, prodrugs that are selectively activated in hypoxic cells or inhibitors of molecular targets involved in hypoxic cell survival (i.e., hypoxia inducible factors HIFs, PI3K/AKT/mTOR pathway, unfolded protein response). While numerous strategies were successful in pre-clinical models, their translation in the clinical practice has been disappointing so far. This therapeutic failure often results from the absence of appropriate stratification of patients that could benefit from targeted interventions. Companion diagnostics may help at different levels of the research and development, and in matching a patient to a specific intervention targeting hypoxia. In this review, we discuss the relative merits of the existing hypoxia biomarkers, their current status and the challenges for their future validation as companion diagnostics adapted to the nature of the intervention.
Collapse
Affiliation(s)
- Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
6
|
Ferda J, Ferdová E, Vítovec M, Glanc D, Mírka H. The imaging of the hypoxic microenvironment in tumorous tissue using PET/CT and PET/MRI. Eur J Radiol 2022; 154:110458. [DOI: 10.1016/j.ejrad.2022.110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/25/2022] [Indexed: 11/03/2022]
|
7
|
Guljaš S, Benšić M, Krivdić Dupan Z, Pavlović O, Krajina V, Pavoković D, Šmit Takač P, Hranić M, Salha T. Dynamic Contrast Enhanced Study in Multiparametric Examination of the Prostate—Can We Make Better Use of It? Tomography 2022; 8:1509-1521. [PMID: 35736872 PMCID: PMC9231365 DOI: 10.3390/tomography8030124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/18/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022] Open
Abstract
We sought to investigate whether quantitative parameters from a dynamic contrast-enhanced study can be used to differentiate cancer from normal tissue and to determine a cut-off value of specific parameters that can predict malignancy more accurately, compared to the obturator internus muscle as a reference tissue. This retrospective study included 56 patients with biopsy proven prostate cancer (PCa) after multiparametric magnetic resonance imaging (mpMRI), with a total of 70 lesions; 39 were located in the peripheral zone, and 31 in the transition zone. The quantitative parameters for all patients were calculated in the detected lesion, morphologically normal prostate tissue and the obturator internus muscle. Increase in the Ktrans value was determined in lesion-to-muscle ratio by 3.974368, which is a cut-off value to differentiate between prostate cancer and normal prostate tissue, with specificity of 72.86% and sensitivity of 91.43%. We introduced a model to detect prostate cancer that combines Ktrans lesion-to-muscle ratio value and iAUC lesion-to-muscle ratio value, which is of higher accuracy compared to individual variables. Based on this model, we identified the optimal cut-off value with 100% sensitivity and 64.28% specificity. The use of quantitative DCE pharmacokinetic parameters compared to the obturator internus muscle as reference tissue leads to higher diagnostic accuracy for prostate cancer detection.
Collapse
Affiliation(s)
- Silva Guljaš
- Clinical Department of Radiology, University Hospital Centre, 31000 Osijek, Croatia; (Z.K.D.); (M.H.)
- Correspondence:
| | - Mirta Benšić
- Department of Mathematics, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Zdravka Krivdić Dupan
- Clinical Department of Radiology, University Hospital Centre, 31000 Osijek, Croatia; (Z.K.D.); (M.H.)
- Department of Radiology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
| | - Oliver Pavlović
- Department of Urology, University Hospital Centre Osijek, 31000 Osijek, Croatia; (O.P.); (V.K.); (D.P.)
| | - Vinko Krajina
- Department of Urology, University Hospital Centre Osijek, 31000 Osijek, Croatia; (O.P.); (V.K.); (D.P.)
| | - Deni Pavoković
- Department of Urology, University Hospital Centre Osijek, 31000 Osijek, Croatia; (O.P.); (V.K.); (D.P.)
| | - Petra Šmit Takač
- Clinical Department of Surgery, Osijek University Hospital Centre, 31000 Osijek, Croatia;
| | - Matija Hranić
- Clinical Department of Radiology, University Hospital Centre, 31000 Osijek, Croatia; (Z.K.D.); (M.H.)
| | - Tamer Salha
- Department of Radiology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia;
- Department of Teleradiology and Artificial Intelligence, Health Centre Osijek-Baranja County, 31000 Osijek, Croatia
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
8
|
Brender JR, Saida Y, Devasahayam N, Krishna MC, Kishimoto S. Hypoxia Imaging As a Guide for Hypoxia-Modulated and Hypoxia-Activated Therapy. Antioxid Redox Signal 2022; 36:144-159. [PMID: 34428981 PMCID: PMC8856011 DOI: 10.1089/ars.2021.0176] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 01/03/2023]
Abstract
Significance: Oxygen imaging techniques, which can probe the spatiotemporal heterogeneity of tumor oxygenation, could be of significant clinical utility in radiation treatment planning and in evaluating the effectiveness of hypoxia-activated prodrugs. To fulfill these goals, oxygen imaging techniques should be noninvasive, quantitative, and capable of serial imaging, as well as having sufficient temporal resolution to detect the dynamics of tumor oxygenation to distinguish regions of chronic and acute hypoxia. Recent Advances: No current technique meets all these requirements, although all have strengths in certain areas. The current status of positron emission tomography (PET)-based hypoxia imaging, oxygen-enhanced magnetic resonance imaging (MRI), 19F MRI, and electron paramagnetic resonance (EPR) oximetry are reviewed along with their strengths and weaknesses for planning hypoxia-guided, intensity-modulated radiation therapy and detecting treatment response for hypoxia-targeted prodrugs. Critical Issues: Spatial and temporal resolution emerges as a major concern for these areas along with specificity and quantitative response. Although multiple oxygen imaging techniques have reached the investigative stage, clinical trials to test the therapeutic effectiveness of hypoxia imaging have been limited. Future Directions: Imaging elements of the redox environment besides oxygen by EPR and hyperpolarized MRI may have a significant impact on our understanding of the basic biology of the reactive oxygen species response and may extend treatment possibilities.
Collapse
Affiliation(s)
- Jeffrey R. Brender
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Yu Saida
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Nallathamby Devasahayam
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Murali C. Krishna
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| | - Shun Kishimoto
- Radiation Biology Branch, Center for Cancer Research, National
Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
9
|
Paudyal R, Grkovski M, Oh JH, Schöder H, Nunez DA, Hatzoglou V, Deasy JO, Humm JL, Lee NY, Shukla-Dave A. Application of Community Detection Algorithm to Investigate the Correlation between Imaging Biomarkers of Tumor Metabolism, Hypoxia, Cellularity, and Perfusion for Precision Radiotherapy in Head and Neck Squamous Cell Carcinomas. Cancers (Basel) 2021; 13:3908. [PMID: 34359810 PMCID: PMC8345739 DOI: 10.3390/cancers13153908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/26/2021] [Accepted: 07/30/2021] [Indexed: 11/17/2022] Open
Abstract
The present study aimed to investigate the correlation at pre-treatment (TX) between quantitative metrics derived from multimodality imaging (MMI), including 18F-FDG-PET/CT, 18F-FMISO-PET/CT, DW- and DCE-MRI, using a community detection algorithm (CDA) in head and neck squamous cell carcinoma (HNSCC) patients. Twenty-three HNSCC patients with 27 metastatic lymph nodes underwent a total of 69 MMI exams at pre-TX. Correlations among quantitative metrics derived from FDG-PET/CT (SUL), FMSIO-PET/CT (K1, k3, TBR, and DV), DW-MRI (ADC, IVIM [D, D*, and f]), and FXR DCE-MRI [Ktrans, ve, and τi]) were investigated using the CDA based on a "spin-glass model" coupled with the Spearman's rank, ρ, analysis. Mean MRI T2 weighted tumor volumes and SULmean values were moderately positively correlated (ρ = 0.48, p = 0.01). ADC and D exhibited a moderate negative correlation with SULmean (ρ ≤ -0.42, p < 0.03 for both). K1 and Ktrans were positively correlated (ρ = 0.48, p = 0.01). In contrast, Ktrans and k3max were negatively correlated (ρ = -0.41, p = 0.03). CDA revealed four communities for 16 metrics interconnected with 33 edges in the network. DV, Ktrans, and K1 had 8, 7, and 6 edges in the network, respectively. After validation in a larger population, the CDA approach may aid in identifying useful biomarkers for developing individual patient care in HNSCC.
Collapse
Affiliation(s)
- Ramesh Paudyal
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - Milan Grkovski
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - Jung Hun Oh
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.S.); (V.H.)
| | - David Aramburu Nunez
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.S.); (V.H.)
| | - Joseph O. Deasy
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - John L. Humm
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
| | - Nancy Y. Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Amita Shukla-Dave
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.P.); (M.G.); (J.H.O.); (D.A.N.); (J.O.D.); (J.L.H.)
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (H.S.); (V.H.)
| |
Collapse
|
10
|
Gertsenshteyn I, Epel B, Barth E, Leoni L, Markiewicz E, Tsai HM, Fan X, Giurcanu M, Bodero D, Zamora M, Sundramoorthy S, Kim H, Freifelder R, Bhuiyan M, Kucharski A, Karczmar G, Kao CM, Halpern H, Chen CT. Improving Tumor Hypoxia Location in 18F-Misonidazole PET with Dynamic Contrast-enhanced MRI Using Quantitative Electron Paramagnetic Resonance Partial Oxygen Pressure Images. Radiol Imaging Cancer 2021; 3:e200104. [PMID: 33817651 PMCID: PMC8011450 DOI: 10.1148/rycan.2021200104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 11/11/2022]
Abstract
Purpose To enhance the spatial accuracy of fluorine 18 (18F) misonidazole (MISO) PET imaging of hypoxia by using dynamic contrast-enhanced (DCE) MR images as a basis for modifying PET images and by using electron paramagnetic resonance (EPR) partial oxygen pressure (pO2) as the reference standard. Materials and Methods Mice (n = 10) with leg-borne MCa4 mammary carcinomas underwent EPR imaging, T2-weighted and DCE MRI, and 18F-MISO PET/CT. Images were registered to the same space for analysis. The thresholds of hypoxia for PET and EPR images were tumor-to-muscle ratios greater than or equal to 2.2 mm Hg and less than or equal to 14 mm Hg, respectively. The Dice similarity coefficient (DSC) and Hausdorff distance (d H ) were used to quantify the three-dimensional overlap of hypoxia between pO2 EPR and 18F-MISO PET images. A training subset (n = 6) was used to calculate optimal DCE MRI weighting coefficients to relate EPR to the PET signal; the group average weights were then applied to all tumors (from six training mice and four test mice). The DSC and d H were calculated before and after DCE MRI-corrected PET images were obtained to quantify the improvement in overlap with EPR pO2 images for measuring tumor hypoxia. Results The means and standard deviations of the DSC and d H between hypoxic regions in original PET and EPR images were 0.35 mm ± 0.23 and 5.70 mm ± 1.7, respectively, for images of all 10 mice. After implementing a preliminary DCE MRI correction to PET data, the DSC increased to 0.86 mm ± 0.18 and the d H decreased to 2.29 mm ± 0.70, showing significant improvement (P < .001) for images of all 10 mice. Specifically, for images of the four independent test mice, the DSC improved with correction from 0.19 ± 0.28 to 0.80 ± 0.29 (P = .02), and the d H improved from 6.40 mm ± 2.5 to 1.95 mm ± 0.63 (P = .01). Conclusion Using EPR information as a reference standard, DCE MRI information can be used to correct 18F-MISO PET information to more accurately reflect areas of hypoxia.Keywords: Animal Studies, Molecular Imaging, Molecular Imaging-Cancer, PET/CT, MR-Dynamic Contrast Enhanced, MR-Imaging, PET/MR, Breast, Oncology, Tumor Mircoenvironment, Electron Paramagnetic ResonanceSupplemental material is available for this article.© RSNA, 2021.
Collapse
Affiliation(s)
- Inna Gertsenshteyn
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Boris Epel
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Eugene Barth
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Lara Leoni
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Erica Markiewicz
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Hsiu-Ming Tsai
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Xiaobing Fan
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Mihai Giurcanu
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Darwin Bodero
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Marta Zamora
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Subramanian Sundramoorthy
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Heejong Kim
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Richard Freifelder
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Mohammed Bhuiyan
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Anna Kucharski
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Gregory Karczmar
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Chien-Min Kao
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Howard Halpern
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| | - Chin-Tu Chen
- From the Department of Radiology (I.G., X.F., H.K., R.F., M.B., A.K., G.K., C.M.K., C.T.C.), National Institutes of Health Center for Electron Paramagnetic Resonance Imaging in Vivo Physiology (I.G., B.E., E.B., D.B., S.S., H.H.), Department of Radiation and Cellular Oncology (I.G., B.E., E.B., D.B., H.H.), Integrated Small Animal Imaging Research Resource (L.L., E.M., H.M.T., X.F., D.B., M.Z., C.M.K., C.T.C.), and Department of Public Health Sciences (M.G.), University of Chicago, 5841 S Maryland Ave, MC-2026, Chicago, IL 60637
| |
Collapse
|
11
|
Carmona-Bozo JC, Manavaki R, Woitek R, Torheim T, Baxter GC, Caracò C, Provenzano E, Graves MJ, Fryer TD, Patterson AJ, Gilbert FJ. Hypoxia and perfusion in breast cancer: simultaneous assessment using PET/MR imaging. Eur Radiol 2021; 31:333-344. [PMID: 32725330 PMCID: PMC7755870 DOI: 10.1007/s00330-020-07067-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/12/2020] [Accepted: 07/03/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Hypoxia is associated with poor prognosis and treatment resistance in breast cancer. However, the temporally variant nature of hypoxia can complicate interpretation of imaging findings. We explored the relationship between hypoxia and vascular function in breast tumours through combined 18F-fluoromisonidazole (18 F-FMISO) PET/MRI, with simultaneous assessment circumventing the effect of temporal variation in hypoxia and perfusion. METHODS Women with histologically confirmed, primary breast cancer underwent a simultaneous 18F-FMISO-PET/MR examination. Tumour hypoxia was assessed using influx rate constant Ki and hypoxic fractions (%HF), while parameters of vascular function (Ktrans, kep, ve, vp) and cellularity (ADC) were derived from dynamic contrast-enhanced (DCE) and diffusion-weighted (DW)-MRI, respectively. Additional correlates included histological subtype, grade and size. Relationships between imaging variables were assessed using Pearson correlation (r). RESULTS Twenty-nine women with 32 lesions were assessed. Hypoxic fractions > 1% were observed in 6/32 (19%) cancers, while 18/32 (56%) tumours showed a %HF of zero. The presence of hypoxia in lesions was independent of histological subtype or grade. Mean tumour Ktrans correlated negatively with Ki (r = - 0.38, p = 0.04) and %HF (r = - 0.33, p = 0.04), though parametric maps exhibited intratumoural heterogeneity with hypoxic regions colocalising with both hypo- and hyperperfused areas. No correlation was observed between ADC and DCE-MRI or PET parameters. %HF correlated positively with lesion size (r = 0.63, p = 0.001). CONCLUSION Hypoxia measured by 18F-FMISO-PET correlated negatively with Ktrans from DCE-MRI, supporting the hypothesis of perfusion-driven hypoxia in breast cancer. Intratumoural hypoxia-perfusion relationships were heterogeneous, suggesting that combined assessment may be needed for disease characterisation, which could be achieved using simultaneous multimodality imaging. KEY POINTS • At the tumour level, hypoxia measured by 18F-FMISO-PET was negatively correlated with perfusion measured by DCE-MRI, which supports the hypothesis of perfusion-driven hypoxia in breast cancer. • No associations were observed between 18F-FMISO-PET parameters and tumour histology or grade, but tumour hypoxic fractions increased with lesion size. • Intratumoural hypoxia-perfusion relationships were heterogeneous, suggesting that the combined hypoxia-perfusion status of tumours may need to be considered for disease characterisation, which can be achieved via simultaneous multimodality imaging as reported here.
Collapse
Affiliation(s)
- Julia C Carmona-Bozo
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Roido Manavaki
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Ramona Woitek
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel 18-20, 1090, Vienna, Austria
| | - Turid Torheim
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Gabrielle C Baxter
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Corradina Caracò
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Elena Provenzano
- Cancer Research UK - Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
- Cambridge Breast Unit, Cambridge University Hospitals NHS Foundation Trust, Box 97, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Martin J Graves
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- MRIS Unit, Cambridge University Hospitals NHS Foundation Trust, Box 162, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Tim D Fryer
- Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, School of Clinical Medicine, University of Cambridge, Box 65, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Andrew J Patterson
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
- MRIS Unit, Cambridge University Hospitals NHS Foundation Trust, Box 162, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK
| | - Fiona J Gilbert
- Department of Radiology, School of Clinical Medicine, University of Cambridge, Box 218, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
12
|
Besson FL, Fernandez B, Faure S, Mercier O, Seferian A, Mignard X, Mussot S, le Pechoux C, Caramella C, Botticella A, Levy A, Parent F, Bulifon S, Montani D, Mitilian D, Fadel E, Planchard D, Besse B, Ghigna-Bellinzoni MR, Comtat C, Lebon V, Durand E. 18F-FDG PET and DCE kinetic modeling and their correlations in primary NSCLC: first voxel-wise correlative analysis of human simultaneous [18F]FDG PET-MRI data. EJNMMI Res 2020; 10:88. [PMID: 32734484 PMCID: PMC7392998 DOI: 10.1186/s13550-020-00671-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVES To decipher the correlations between PET and DCE kinetic parameters in non-small-cell lung cancer (NSCLC), by using voxel-wise analysis of dynamic simultaneous [18F]FDG PET-MRI. MATERIAL AND METHODS Fourteen treatment-naïve patients with biopsy-proven NSCLC prospectively underwent a 1-h dynamic [18F]FDG thoracic PET-MRI scan including DCE. The PET and DCE data were normalized to their corresponding T1-weighted MR morphological space, and tumors were masked semi-automatically. Voxel-wise parametric maps of PET and DCE kinetic parameters were computed by fitting the dynamic PET and DCE tumor data to the Sokoloff and Extended Tofts models respectively, by using in-house developed procedures. Curve-fitting errors were assessed by computing the relative root mean square error (rRMSE) of the estimated PET and DCE signals at the voxel level. For each tumor, Spearman correlation coefficients (rs) between all the pairs of PET and DCE kinetic parameters were estimated on a voxel-wise basis, along with their respective bootstrapped 95% confidence intervals (n = 1000 iterations). RESULTS Curve-fitting metrics provided fit errors under 20% for almost 90% of the PET voxels (median rRMSE = 10.3, interquartile ranges IQR = 8.1; 14.3), whereas 73.3% of the DCE voxels showed fit errors under 45% (median rRMSE = 31.8%, IQR = 22.4; 46.6). The PET-PET, DCE-DCE, and PET-DCE voxel-wise correlations varied according to individual tumor behaviors. Beyond this wide variability, the PET-PET and DCE-DCE correlations were mainly high (absolute rs values > 0.7), whereas the PET-DCE correlations were mainly low to moderate (absolute rs values < 0.7). Half the tumors showed a hypometabolism with low perfused/vascularized profile, a hallmark of hypoxia, and tumor aggressiveness. CONCLUSION A dynamic "one-stop shop" procedure applied to NSCLC is technically feasible in clinical practice. PET and DCE kinetic parameters assessed simultaneously are not highly correlated in NSCLC, and these correlations showed a wide variability among tumors and patients. These results tend to suggest that PET and DCE kinetic parameters might provide complementary information. In the future, this might make PET-MRI a unique tool to characterize the individual tumor biological behavior in NSCLC.
Collapse
Affiliation(s)
- Florent L Besson
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMAPs, 91401, Orsay, France.
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270, Le Kremlin-Bicêtre, France.
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France.
| | | | - Sylvain Faure
- Laboratoire de Mathématiques d'Orsay, CNRS, Université Paris-Saclay, 91405, Orsay, France
| | - Olaf Mercier
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Andrei Seferian
- Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, 94270, Le Kremlin-Bicêtre, France
- Inserm UMR_S999, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Xavier Mignard
- Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, 94270, Le Kremlin-Bicêtre, France
| | - Sacha Mussot
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Cecile le Pechoux
- Department of Radiation Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Caroline Caramella
- Department of Radiology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Angela Botticella
- Department of Radiation Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Antonin Levy
- Department of Radiation Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Florence Parent
- Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, 94270, Le Kremlin-Bicêtre, France
- Inserm UMR_S999, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Sophie Bulifon
- Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, 94270, Le Kremlin-Bicêtre, France
- Inserm UMR_S999, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - David Montani
- Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, 94270, Le Kremlin-Bicêtre, France
- Inserm UMR_S999, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Delphine Mitilian
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - Elie Fadel
- Department of Thoracic and Vascular Surgery and Heart-Lung Transplantation, Marie Lannelongue Hospital, 92350, Le Plessis Robinson, France
| | - David Planchard
- Department of Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | - Benjamin Besse
- Department of Oncology, Institut d'Oncologie Thoracique (IOT), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Claude Comtat
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMAPs, 91401, Orsay, France
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Vincent Lebon
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMAPs, 91401, Orsay, France
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Emmanuel Durand
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMAPs, 91401, Orsay, France
- Department of Biophysics and Nuclear Medicine-Molecular Imaging, Hôpitaux Universitaires Paris-Saclay, Assistance Publique-Hôpitaux de Paris, CHU Bicêtre, 94270, Le Kremlin-Bicêtre, France
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
13
|
Combination of DCE-MRI and DWI in Predicting the Treatment Effect of Concurrent Chemoradiotherapy in Esophageal Carcinoma. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2576563. [PMID: 32626736 PMCID: PMC7315287 DOI: 10.1155/2020/2576563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 12/26/2019] [Accepted: 01/30/2020] [Indexed: 11/17/2022]
Abstract
Background Concurrent chemoradiotherapy (CCRT) is the main treatment for esophageal cancer, but the response to treatment varies from individual to individual. MR imaging methods, such as diffusion-weighted (DW) MRI and the use of dynamic contrast-enhanced (DCE) MRI, have the potential to provide additional biomarkers that could evaluate the effect of CCRT in patients with esophageal carcinoma. Materials and Methods Fifty-six patients with esophageal carcinoma, verified by histopathology, underwent MRI examination before and at midtreatment (4th week, radiotherapy 30-40 Gy) using the Siemens 3.0 T MR System. Parameter maps of apparent diffusion coefficient (ADC), and DCE maps of volume transfer constant (K rans), rate contrast (k ep), and extracellular fluid space (v e), were computed using a Siemens Company Multimodality Workplace (MMWP) model. Comparison of histogram parameters and their diagnostic performance was determined using the Mann-Whitney U test and receiver operating characteristic (ROC) analysis. Results 56 patient MRI scans were available for analysis at baseline and at the third week, respectively. Pretreatment K rans, pretreatment k ep, pretreatment ADC (P < 0.05), and during-treatment K rans (P < 0.05) and ΔK rans and ΔADC (P < 0.05) were significantly different after CCRT. Based on the binary logistic model, the ROC analysis demonstrated that the combined predictors demonstrated a high diagnostic performance with an AUC of 0.939. The sensitivity and specificity were 98.6% and 73.8%, respectively. Conclusion The combination of DCE and DWI can be used as an early biomarker in the prediction of the effect of CCRT three weeks after treatment in esophageal carcinoma.
Collapse
|
14
|
Wiedenmann N, Grosu AL, Büchert M, Rischke HC, Ruf J, Bielak L, Majerus L, Rühle A, Bamberg F, Baltas D, Hennig J, Mix M, Bock M, Nicolay NH. The utility of multiparametric MRI to characterize hypoxic tumor subvolumes in comparison to FMISO PET/CT. Consequences for diagnosis and chemoradiation treatment planning in head and neck cancer. Radiother Oncol 2020; 150:128-135. [PMID: 32544609 DOI: 10.1016/j.radonc.2020.06.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/02/2020] [Accepted: 06/10/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND PURPOSE Hypoxia is an essential metabolic marker that determines chemo- and radiation resistance in head-and-neck squamous cell carcinoma (HNSCC) patients. Our exploratory analysis aimed to identify multiparametric MRI (mpMRI) parameters linked to hypoxia that might be used as surrogate for [18F]FMISO-PET in diagnosis and chemoradiation treatment (CRT) of HNSCC. MATERIALS AND METHODS 21 patients undergoing definitive CRT for HNSCC were prospectively imaged with serial [18F]FMISO-PET and 3 Tesla mpMRI for T1- and T2-weighted and dynamic contrast-enhanced perfusion and diffusion-weighted measurements (ktrans, ve, kep, ADC) in weeks 0, 2 and 5 and FDG-PET in week 0. [18F]FMISO-PET-derived hypoxic subvolumes (HSV) and complementary non-hypoxic subvolumes (nonHSV) were created for tumor and lymph nodes and projected on the mpMRI scans after PET/MRI co-registration. MpMRI and [18F]FMISO-PET parameters within HSVs and nonHSVs were statistically compared. RESULTS FMISO-PET-based HSVs of the primary tumors on MRI were characterized by lower ADC at all time points (p = 0.012 at baseline; p = 0.015 in week 2) and reduced interstitial space volume fraction ve and perfusion ktrans at baseline (p = 0.006, p = 0.047) compared to nonHSVs. Hypoxic lymph nodes were characterized by significantly lower ADC values at baseline (p = 0.039), but not at later time points and a reduction in ktrans-based perfusion at week 2 (p = 0.018). CONCLUSION MpMRI parameters differ significantly between hypoxic and non-hypoxic tumor regions, defined on FMISO-PET/CT as gold standard and might represent surrogate markers for tumor hypoxia. These findings suggest that mpMRI may be useful in the future as a surrogate modality for hypoxia imaging in order to personalize CRT.
Collapse
Affiliation(s)
- Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Büchert
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans C Rischke
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bielak
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Liette Majerus
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Bamberg
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Hennig
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Bock
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner site Freiburg, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
15
|
Butler M, Perperidis A, Zahra JLM, Silva N, Averkiou M, Duncan WC, McNeilly A, Sboros V. Differentiation of Vascular Characteristics Using Contrast-Enhanced Ultrasound Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:2444-2455. [PMID: 31208880 DOI: 10.1016/j.ultrasmedbio.2019.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 05/09/2023]
Abstract
Ultrasound contrast imaging has been used to assess tumour growth and regression by assessing the flow through the macro- and micro-vasculature. Our aim was to differentiate the blood kinetics of vessels such as veins, arteries and microvasculature within the limits of the spatial resolution of contrast-enhanced ultrasound imaging. The highly vascularised ovine ovary was used as a biological model. Perfusion of the ovary with SonoVue was recorded with a Philips iU22 scanner in contrast imaging mode. One ewe was treated with prostaglandin to induce vascular regression. Time-intensity curves (TIC) for different regions of interest were obtained, a lognormal model was fitted and flow parameters calculated. Parametric maps of the whole imaging plane were generated for 2 × 2 pixel regions of interest. Further analysis of TICs from selected locations helped specify parameters associated with differentiation into four categories of vessels (arteries, veins, medium-sized vessels and micro-vessels). Time-dependent parameters were associated with large veins, whereas intensity-dependent parameters were associated with large arteries. Further development may enable automation of the technique as an efficient way of monitoring vessel distributions in a clinical setting using currently available scanners.
Collapse
Affiliation(s)
- Mairead Butler
- Heriot-Watt University, Institute of Biochemistry, Biological Physics and Bio Engineering, Riccarton, Edinburgh, UK.
| | - Antonios Perperidis
- Heriot-Watt University, Institute of Signals, Sensors and Systems, Riccarton, Edinburgh, UK
| | | | - Nadia Silva
- Centre for Marine Sciences, University of Algarve Faro, Portugal
| | - Michalakis Averkiou
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
| | - W Colin Duncan
- Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Alan McNeilly
- Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Vassilis Sboros
- Heriot-Watt University, Institute of Biochemistry, Biological Physics and Bio Engineering, Riccarton, Edinburgh, UK
| |
Collapse
|
16
|
Eckert F, Zwirner K, Boeke S, Thorwarth D, Zips D, Huber SM. Rationale for Combining Radiotherapy and Immune Checkpoint Inhibition for Patients With Hypoxic Tumors. Front Immunol 2019; 10:407. [PMID: 30930892 PMCID: PMC6423917 DOI: 10.3389/fimmu.2019.00407] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 02/15/2019] [Indexed: 12/19/2022] Open
Abstract
In order to compensate for the increased oxygen consumption in growing tumors, tumors need angiogenesis and vasculogenesis to increase the supply. Insufficiency in this process or in the microcirculation leads to hypoxic tumor areas with a significantly reduced pO2, which in turn leads to alterations in the biology of cancer cells as well as in the tumor microenvironment. Cancer cells develop more aggressive phenotypes, stem cell features and are more prone to metastasis formation and migration. In addition, intratumoral hypoxia confers therapy resistance, specifically radioresistance. Reactive oxygen species are crucial in fixing DNA breaks after ionizing radiation. Thus, hypoxic tumor cells show a two- to threefold increase in radioresistance. The microenvironment is enriched with chemokines (e.g., SDF-1) and growth factors (e.g., TGFβ) additionally reducing radiosensitivity. During recent years hypoxia has also been identified as a major factor for immune suppression in the tumor microenvironment. Hypoxic tumors show increased numbers of myeloid derived suppressor cells (MDSCs) as well as regulatory T cells (Tregs) and decreased infiltration and activation of cytotoxic T cells. The combination of radiotherapy with immune checkpoint inhibition is on the rise in the treatment of metastatic cancer patients, but is also tested in multiple curative treatment settings. There is a strong rationale for synergistic effects, such as increased T cell infiltration in irradiated tumors and mitigation of radiation-induced immunosuppressive mechanisms such as PD-L1 upregulation by immune checkpoint inhibition. Given the worse prognosis of patients with hypoxic tumors due to local therapy resistance but also increased rate of distant metastases and the strong immune suppression induced by hypoxia, we hypothesize that the subgroup of patients with hypoxic tumors might be of special interest for combining immune checkpoint inhibition with radiotherapy.
Collapse
Affiliation(s)
- Franziska Eckert
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Zwirner
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Simon Boeke
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Consortium (DKTK) Partnersite Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan M. Huber
- Department of Radiation Oncology, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
17
|
Bonnitcha P, Grieve S, Figtree G. Clinical imaging of hypoxia: Current status and future directions. Free Radic Biol Med 2018; 126:296-312. [PMID: 30130569 DOI: 10.1016/j.freeradbiomed.2018.08.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 12/20/2022]
Abstract
Tissue hypoxia is a key feature of many important causes of morbidity and mortality. In pathologies such as stroke, peripheral vascular disease and ischaemic heart disease, hypoxia is largely a consequence of low blood flow induced ischaemia, hence perfusion imaging is often used as a surrogate for hypoxia to guide clinical diagnosis and treatment. Importantly, ischaemia and hypoxia are not synonymous conditions as it is not universally true that well perfused tissues are normoxic or that poorly perfused tissues are hypoxic. In pathologies such as cancer, for instance, perfusion imaging and oxygen concentration are less well correlated, and oxygen concentration is independently correlated to radiotherapy response and overall treatment outcomes. In addition, the progression of many diseases is intricately related to maladaptive responses to the hypoxia itself. Thus there is potentially great clinical and scientific utility in direct measurements of tissue oxygenation. Despite this, imaging assessment of hypoxia in patients is rarely performed in clinical settings. This review summarises some of the current methods used to clinically evaluate hypoxia, the barriers to the routine use of these methods and the newer agents and techniques being explored for the assessment of hypoxia in pathological processes.
Collapse
Affiliation(s)
- Paul Bonnitcha
- Northern and Central Clinical Schools, Faculty of Medicine, Sydney University, Sydney, NSW 2006, Australia; Chemical Pathology Department, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia.
| | - Stuart Grieve
- Sydney Translational Imaging Laboratory, Heart Research Institute, Charles Perkins Centre and Sydney Medical School, University of Sydney, NSW 2050, Australia
| | - Gemma Figtree
- Kolling Institute of Medical Research, University of Sydney, St Leonards, New South Wales 2065, Australia; Cardiology Department, Royal North Shore Hospital, St Leonards, New South Wales 2065, Australia
| |
Collapse
|
18
|
Wiedenmann N, Bunea H, Rischke HC, Bunea A, Majerus L, Bielak L, Protopopov A, Ludwig U, Büchert M, Stoykow C, Nicolay NH, Weber WA, Mix M, Meyer PT, Hennig J, Bock M, Grosu AL. Effect of radiochemotherapy on T2* MRI in HNSCC and its relation to FMISO PET derived hypoxia and FDG PET. Radiat Oncol 2018; 13:159. [PMID: 30157883 PMCID: PMC6114038 DOI: 10.1186/s13014-018-1103-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/17/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND To assess the effect of radiochemotherapy (RCT) on proposed tumour hypoxia marker transverse relaxation time (T2*) and to analyse the relation between T2* and 18F-misonidazole PET/CT (FMISO-PET) and 18F-fluorodeoxyglucose PET/CT (FDG-PET). METHODS Ten patients undergoing definitive RCT for squamous cell head-and-neck cancer (HNSCC) received repeat FMISO- and 3 Tesla T2*-weighted MRI at weeks 0, 2 and 5 during treatment and FDG-PET at baseline. Gross tumour volumes (GTV) of tumour (T), lymph nodes (LN) and hypoxic subvolumes (HSV, based on FMISO-PET) and complementary non-hypoxic subvolumes (nonHSV) were generated. Mean values for T2* and SUVmean FDG were determined. RESULTS During RCT, marked reduction of tumour hypoxia on FMISO-PET was observed (T, LN), while mean T2* did not change significantly. At baseline, mean T2* values within HSV-T (15 ± 5 ms) were smaller compared to nonHSV-T (18 ± 3 ms; p = 0.051), whereas FDG SUVmean (12 ± 6) was significantly higher for HSV-T (12 ± 6) than for nonHSV-T (6 ± 3; p = 0.026) and higher for HSV-LN (10 ± 4) than for nonHSV-LN (5 ± 2; p ≤ 0.011). Correlation between FMISO PET and FDG PET was higher than between FMSIO PET and T2* (R2 for GTV-T (FMISO/FDG) = 0.81, R2 for GTV-T (FMISO/T2*) = 0.32). CONCLUSIONS Marked reduction of tumour hypoxia between week 0, 2 and 5 found on FMISO PET was not accompanied by a significant T2*change within GTVs over time. These results suggest a relation between tumour oxygenation status and T2* at baseline, but no simple correlation over time. Therefore, caution is warranted when using T2* as a substitute for FMISO-PET to monitor tumour hypoxia during RCT in HNSCC patients. TRIAL REGISTRATION DRKS, DRKS00003830 . Registered 23.04.2012.
Collapse
Affiliation(s)
- Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany. .,German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Hatice Bunea
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans C Rischke
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrei Bunea
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Liette Majerus
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Bielak
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexey Protopopov
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ute Ludwig
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Büchert
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christian Stoykow
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfgang A Weber
- Clinic for Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jürgen Hennig
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Bock
- Department of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
19
|
Kargar S, Borisch EA, Froemming AT, Kawashima A, Mynderse LA, Stinson EG, Trzasko JD, Riederer SJ. Robust and efficient pharmacokinetic parameter non-linear least squares estimation for dynamic contrast enhanced MRI of the prostate. Magn Reson Imaging 2017; 48:50-61. [PMID: 29278764 DOI: 10.1016/j.mri.2017.12.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Revised: 12/09/2017] [Accepted: 12/21/2017] [Indexed: 12/15/2022]
Abstract
PURPOSE To describe an efficient numerical optimization technique using non-linear least squares to estimate perfusion parameters for the Tofts and extended Tofts models from dynamic contrast enhanced (DCE) MRI data and apply the technique to prostate cancer. METHODS Parameters were estimated by fitting the two Tofts-based perfusion models to the acquired data via non-linear least squares. We apply Variable Projection (VP) to convert the fitting problem from a multi-dimensional to a one-dimensional line search to improve computational efficiency and robustness. Using simulation and DCE-MRI studies in twenty patients with suspected prostate cancer, the VP-based solver was compared against the traditional Levenberg-Marquardt (LM) strategy for accuracy, noise amplification, robustness to converge, and computation time. RESULTS The simulation demonstrated that VP and LM were both accurate in that the medians closely matched assumed values across typical signal to noise ratio (SNR) levels for both Tofts models. VP and LM showed similar noise sensitivity. Studies using the patient data showed that the VP method reliably converged and matched results from LM with approximate 3× and 2× reductions in computation time for the standard (two-parameter) and extended (three-parameter) Tofts models. While LM failed to converge in 14% of the patient data, VP converged in the ideal 100%. CONCLUSION The VP-based method for non-linear least squares estimation of perfusion parameters for prostate MRI is equivalent in accuracy and robustness to noise, while being more reliably (100%) convergent and computationally about 3× (TM) and 2× (ETM) faster than the LM-based method.
Collapse
Affiliation(s)
- Soudabeh Kargar
- Biomedical Engineering and Physiology Program, Mayo Graduate School, Rochester, MN, United States; Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Eric A Borisch
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Adam T Froemming
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Akira Kawashima
- Department of Radiology, Mayo Clinic, Scottsdale, AZ, United States
| | - Lance A Mynderse
- Department of Urology, Mayo Clinic, Rochester, MN, United States
| | - Eric G Stinson
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Joshua D Trzasko
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Stephen J Riederer
- Biomedical Engineering and Physiology Program, Mayo Graduate School, Rochester, MN, United States; Department of Radiology, Mayo Clinic, Rochester, MN, United States.
| |
Collapse
|
20
|
Daniel M, Andrzejewski P, Sturdza A, Majercakova K, Baltzer P, Pinker K, Wadsak W, Mitterhauser M, Pötter R, Georg P, Helbich T, Georg D. Impact of hybrid PET/MR technology on multiparametric imaging and treatment response assessment of cervix cancer. Radiother Oncol 2017; 125:420-425. [DOI: 10.1016/j.radonc.2017.10.036] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/27/2017] [Accepted: 10/28/2017] [Indexed: 12/12/2022]
|
21
|
|
22
|
Thorwarth D, Wack LJ, Mönnich D. Hypoxia PET imaging techniques: data acquisition and analysis. Clin Transl Imaging 2017. [DOI: 10.1007/s40336-017-0250-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Puri T, Greenhalgh TA, Wilson JM, Franklin J, Wang LM, Strauss V, Cunningham C, Partridge M, Maughan T. [ 18F]Fluoromisonidazole PET in rectal cancer. EJNMMI Res 2017; 7:78. [PMID: 28933018 PMCID: PMC5607050 DOI: 10.1186/s13550-017-0324-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is an increasing interest in developing predictive biomarkers of tissue hypoxia using functional imaging for personalised radiotherapy in patients with rectal cancer that are considered for neoadjuvant chemoradiotherapy (CRT). The study explores [18F]fluoromisonidazole ([18F]FMISO) positron emission tomography (PET) scans for predicting clinical response in rectal cancer patients receiving neoadjuvant CRT. METHODS Patients with biopsy-proven rectal adenocarcinoma were imaged at 0-45 min, 2 and 4 h, at baseline and after 8-10 fractions of CRT (week 2). The first 6 patients did not receive an enema (the non-enema group) and the last 4 patients received an enema before PET-CT scan (the enema group). [18F]FMISO production failed on 2 occasions. Static PET images at 4 h were analysed using tumour-to-muscle (T:M) SUVmax and tumour-to-blood (T:B) SUVmax. The 0-45 min dynamic PET scans were analysed using Casciari model to report hypoxia and perfusion. Akaike information criteria (AIC) were used to compare data fittings for different pharmacokinetic models. Pathological tumour regression grade was scored using American Joint Committee on Cancer (AJCC) 7.0. Shapiro-Wilk test was used to evaluate the normality of the data. RESULTS Five out of eleven (5/11) patients were classed as good responders (AJCC 0/1 or good clinical response) and 6/11 as poor responders (AJCC 2/3 or poor clinical response). The median T:M SUVmax was 2.14 (IQR 0.58) at baseline and 1.30 (IQR 0.19) at week 2, and the corresponding median tumour hypoxia volume was 1.08 (IQR 1.31) cm3 and 0 (IQR 0.15) cm3, respectively. The median T:B SUVmax was 2.46 (IQR 1.50) at baseline and 1.61 (IQR 0.14) at week 2, and the corresponding median tumour hypoxia volume was 5.68 (IQR 5.86) cm3 and 0.76 (IQR 0.78) cm3, respectively. For 0-45 min tumour modelling, the median hypoxia was 0.92 (IQR 0.41) min-1 at baseline and 0.70 (IQR 0.10) min-1 at week 2. The median perfusion was 4.10 (IQR 1.71) ml g-1 min-1 at baseline and 2.48 (IQR 3.62) ml g-1 min-1 at week 2. In 9/11 patients with both PET scans, tumour perfusion decreased in non-responders and increased in responders except in one patient. None of the changes in other PET parameters showed any clear trend with clinical outcome. CONCLUSIONS This pilot study with small number of datasets revealed significant challenges in delivery and interpretation of [18F]FMISO PET scans of rectal cancer. There are two principal problems namely spill-in from non-tumour tracer activity from rectal and bladder contents. Emphasis should be made on reducing spill-in effects from the bladder to improve data quality. This preliminary study has shown fundamental difficulties in the interpretation of [18F]FMISO PET scans for rectal cancer, limiting its clinical applicability.
Collapse
Affiliation(s)
- Tanuj Puri
- CRUK/MRC Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Tessa A. Greenhalgh
- CRUK/MRC Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ UK
| | - James M. Wilson
- CRUK/MRC Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Jamie Franklin
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Lia Mun Wang
- Department of Cellular Pathology, John Radcliffe Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Present address: Department of Laboratory Medicine, Changi General Hospital, 2 Simei Street 3, Singapore, Singapore
| | - Victoria Strauss
- Centre for Statistics in Medicine, Oxford Clinical Trial Research Unit, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Chris Cunningham
- Department of Colorectal Surgery, Cancer Centre, Churchill Hospital, Oxford, University Hospitals NHS Foundation Trust, Oxford, UK
| | - Mike Partridge
- CRUK/MRC Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ UK
| | - Tim Maughan
- CRUK/MRC Oxford Institute of Radiation Oncology, Department of Oncology, University of Oxford, Old Road Campus Research Building, Off Roosevelt Drive, Oxford, OX3 7DQ UK
| |
Collapse
|
24
|
Thorwarth D. Biologically adapted radiation therapy. Z Med Phys 2017; 28:177-183. [PMID: 28869163 DOI: 10.1016/j.zemedi.2017.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 01/05/2023]
Abstract
The aim of biologically adapted radiotherapy (RT) is to shape or paint the prescribed radiation dose according to biological properties of the tumor in order to increase local control rates in the future. Human tumors are known to present with an extremely heterogeneous tissue architecture leading to highly variable local cell densities and chaotic vascular structures leading to tumor hypoxia and regions of increased radiation resistance. The goal of biologically adapted RT or dose painting is to individually adapt the radiation dose to biological features of the tumor as non-invasively assessed with functional imaging in order to overcome increased radiation resistance. This article discusses the whole development chain of biologically adapted RT from radio-biologically relevant processes, functional imaging techniques to visualize tumor biology non-invasively and radiation prescription functions to the implementation of biologically adapted RT in clinical practice.
Collapse
Affiliation(s)
- Daniela Thorwarth
- Sektion Biomedizinische Physik, Universitätsklinikum für Radioonkologie, Eberhard Karls Universität Tübingen, Germany.
| |
Collapse
|
25
|
Seith F, Forschner A, Schmidt H, Pfannenberg C, Gückel B, Nikolaou K, la Fougère C, Garbe C, Schwenzer N. 18F-FDG-PET detects complete response to PD1-therapy in melanoma patients two weeks after therapy start. Eur J Nucl Med Mol Imaging 2017; 45:95-101. [PMID: 28831583 DOI: 10.1007/s00259-017-3813-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/15/2017] [Indexed: 12/29/2022]
Abstract
PURPOSE The aim of the study was to evaluate if 18F-FDG-PET has the potential to detect complete responders to PD1-therapy in patients with unresectable metastasized melanoma two weeks after therapy initiation. METHODS Between September 2014 and May 2016, ten patients (four females; 65 ± 12 y) received a whole-body 18F-FDG-PET/MRI examination at three time points: Before therapy start (t0, base-line), two weeks (t1, study examination) and three months after treatment initiation (t2, reference standard). Therapy response was assessed with PET response criteria in solid tumors (PERCIST). Time to progression and overall survival (OS) were obtained for all patients. RESULTS Three patients with partial metabolic response in PET at t1 turned out to have complete response at t2. No tumor relapse was observed in those patients so far (observation period: 265, 511 and 728 days, respectively). At t2, progressive metabolic disease (PMD) was seen in six patients from whom four showed PMD and two showed stable metabolic disease (SMD) at t1. OS in patients with PMD at t2 varied between 148 and 814 days. SMD at both t1 and t2 was seen in one patient, tumor progress was observed after 308 days. CONCLUSION Our study indicates that whole-body 18F-FDG-PET might be able to reliably identify complete responders to PD1-therapy as early as two weeks after therapy initiation in stage IV melanoma patients. This might help to shorten therapy regimes and avoid unnecessary side effects in the future.
Collapse
Affiliation(s)
- Ferdinand Seith
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany.
| | - Andrea Forschner
- Department of Dermatology, Eberhard Karls University, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Holger Schmidt
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany
| | - Christina Pfannenberg
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany
| | - Brigitte Gückel
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany
| | - Konstantin Nikolaou
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Christian la Fougère
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Eberhard Karls University, Otfried-Mueller-Strasse 14, 72076, Tuebingen, Germany
| | - Claus Garbe
- Department of Dermatology, Eberhard Karls University, Liebermeisterstrasse 25, 72076, Tuebingen, Germany
| | - Nina Schwenzer
- Diagnostic and Interventional Radiology, Department of Radiology, Eberhard Karls University, Hoppe-Seyler-Strasse 3, 72076, Tuebingen, Germany
| |
Collapse
|
26
|
Peerlings J, Van De Voorde L, Mitea C, Larue R, Yaromina A, Sandeleanu S, Spiegelberg L, Dubois L, Lambin P, Mottaghy FM. Hypoxia and hypoxia response-associated molecular markers in esophageal cancer: A systematic review. Methods 2017; 130:51-62. [PMID: 28705470 DOI: 10.1016/j.ymeth.2017.07.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 12/22/2022] Open
Abstract
PURPOSE In this systematic review, the existing evidence of available hypoxia-associated molecular response biomarkers in esophageal cancer (EC) patients is summarized and set into the context of the role of hypoxia in the prediction of esophageal cancer, treatment response and treatment outcome. METHODS A systematic literature search was performed in Web of Science, MEDLINE, and PubMed databases using the keywords: hypoxia, esophagus, cancer, treatment outcome and treatment response. Eligible publications were independently evaluated by two reviewers. In total, 22 out of 419 records were included for systematic review. The described search strategy was applied weekly, with the last update being performed on April 3rd, 2017. RESULTS In esophageal cancer, several (non-)invasive biomarkers for hypoxia could be identified. Independent prognostic factors for treatment response include HIF-1α, CA IX, GLUT-1 overexpression and elevated uptake of the PET-tracer 18F-fluoroerythronitroimidazole (18F-FETNIM). Hypoxia-associated molecular responses represents a clinically relevant phenomenon in esophageal cancer and detection of elevated levels of hypoxia-associated biomarkers and tends to be associated with poor treatment outcome (i.e., overall survival, disease-free survival, complete response and local control). CONCLUSION Evaluation of tumor micro-environmental conditions, such as intratumoral hypoxia, is important to predict treatment outcome and efficacy. Promising non-invasive imaging-techniques have been suggested to assess tumor hypoxia and hypoxia-associated molecular responses. However, extensive validation in EC is lacking. Hypoxia-associated markers that are independent prognostic factors could potentially provide targets for novel treatment strategies to improve treatment outcome. For personalized hypoxia-guided treatment, safe and reliable makers for tumor hypoxia are needed to select suitable patients.
Collapse
Affiliation(s)
- Jurgen Peerlings
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Lien Van De Voorde
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Cristina Mitea
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ruben Larue
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ala Yaromina
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Sebastian Sandeleanu
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Linda Spiegelberg
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ludwig Dubois
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Philippe Lambin
- MAASTRO Clinic, Department of Radiation Oncology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, Maastricht, The Netherlands; Department of Nuclear Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| |
Collapse
|
27
|
Challapalli A, Carroll L, Aboagye EO. Molecular mechanisms of hypoxia in cancer. Clin Transl Imaging 2017; 5:225-253. [PMID: 28596947 PMCID: PMC5437135 DOI: 10.1007/s40336-017-0231-1] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 04/21/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE Hypoxia is a condition of insufficient oxygen to support metabolism which occurs when the vascular supply is interrupted, or when a tumour outgrows its vascular supply. It is a negative prognostic factor due to its association with an aggressive tumour phenotype and therapeutic resistance. This review provides an overview of hypoxia imaging with Positron emission tomography (PET), with an emphasis on the biological relevance, mechanism of action, highlighting advantages, and limitations of the currently available hypoxia radiotracers. METHODS A comprehensive PubMed literature search was performed, identifying articles relating to biological significance and measurement of hypoxia, MRI methods, and PET imaging of hypoxia in preclinical and clinical settings, up to December 2016. RESULTS A variety of approaches have been explored over the years for detecting and monitoring changes in tumour hypoxia, including regional measurements with oxygen electrodes placed under CT guidance, MRI methods that measure either oxygenation or lactate production consequent to hypoxia, different nuclear medicine approaches that utilise imaging agents the accumulation of which is inversely related to oxygen tension, and optical methods. The advantages and disadvantages of these approaches are reviewed, along with individual strategies for validating different imaging methods. PET is the preferred method for imaging tumour hypoxia due to its high specificity and sensitivity to probe physiological processes in vivo, as well as the ability to provide information about intracellular oxygenation levels. CONCLUSION Even though hypoxia could have significant prognostic and predictive value in the clinic, the best method for hypoxia assessment has in our opinion not been realised.
Collapse
Affiliation(s)
- Amarnath Challapalli
- Department of Clinical Oncology, Bristol Cancer Institute, Horfield Road, Bristol, United Kingdom
| | - Laurence Carroll
- Department of Surgery and Cancer, Imperial College, GN1, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W120NN United Kingdom
| | - Eric O. Aboagye
- Department of Surgery and Cancer, Imperial College, GN1, Commonwealth Building, Hammersmith Hospital, Du Cane Road, London, W120NN United Kingdom
| |
Collapse
|