1
|
Ye Y, Cao Z. Glucose Metabolism and Glucose Transporters in Head and Neck Squamous Cell Carcinoma. Cancer Invest 2024:1-18. [PMID: 39324504 DOI: 10.1080/07357907.2024.2407424] [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: 05/11/2024] [Accepted: 09/18/2024] [Indexed: 09/27/2024]
Abstract
Head and neck squamous cell carcinoma ranks seventh globally in malignancy prevalence, with persistent high mortality rates despite treatment advancements. Glucose, pivotal in cancer metabolism via the Warburg effect, enters cells via glucose transporters, notably GLUT proteins. Glycolysis, aerobic oxidation, and the pentose phosphate pathway in glucose metabolism significantly impact HNSCC progression. HNSCC exhibits elevated expression of glucose metabolism enzymes and GLUT proteins, correlating with prognosis. Heterogeneity in HNSCC yields varied metabolic profiles, influenced by factors like HPV status and disease stage. This review highlights glucose metabolism's role and potential as therapeutic targets and cancer imaging tracers in HNSCC.
Collapse
Affiliation(s)
- Yanyan Ye
- Department of Otolaryngology, Shulan (Hangzhou) Hospital, affiliated to Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Zaizai Cao
- Department of Otolaryngology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
2
|
Rühle A, Nicolay NH. [Hypoxia-based de-escalation of radiochemotherapy in patients with human papillomavirus-related oropharyngeal carcinoma]. Strahlenther Onkol 2024; 200:453-456. [PMID: 38396139 DOI: 10.1007/s00066-024-02215-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 02/25/2024]
Affiliation(s)
- Alexander Rühle
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig, Stephanstraße 9a, 04103, Leipzig, Deutschland.
- Arbeitsgruppe junge DEGRO der Deutschen Gesellschaft für Radioonkologie e. V. (DEGRO), Berlin, Deutschland.
- Mitteldeutsches Krebszentrum (CCCG), Partnerstandort Leipzig, Leipzig, Deutschland.
| | - Nils H Nicolay
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Leipzig, Stephanstraße 9a, 04103, Leipzig, Deutschland
- Mitteldeutsches Krebszentrum (CCCG), Partnerstandort Leipzig, Leipzig, Deutschland
| |
Collapse
|
3
|
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
|
4
|
Gouel P, Callonnec F, Obongo-Anga FR, Bohn P, Lévêque E, Gensanne D, Hapdey S, Modzelewski R, Vera P, Thureau S. Quantitative MRI to Characterize Hypoxic Tumors in Comparison to FMISO PET/CT for Radiotherapy in Oropharynx Cancers. Cancers (Basel) 2023; 15:cancers15061918. [PMID: 36980806 PMCID: PMC10047588 DOI: 10.3390/cancers15061918] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Intratumoral hypoxia is associated with a poor prognosis and poor response to treatment in head and neck cancers. Its identification would allow for increasing the radiation dose to hypoxic tumor subvolumes. 18F-FMISO PET imaging is the gold standard; however, quantitative multiparametric MRI could show the presence of intratumoral hypoxia. Thus, 16 patients were prospectively included and underwent 18F-FDG PET/CT, 18F-FMISO PET/CT, and multiparametric quantitative MRI (DCE, diffusion and relaxometry T1 and T2 techniques) in the same position before treatment. PET and MRI sub-volumes were segmented and classified as hypoxic or non-hypoxic volumes to compare quantitative MRI parameters between normoxic and hypoxic volumes. In total, 13 patients had hypoxic lesions. The Dice, Jaccard, and overlap fraction similarity indices were 0.43, 0.28, and 0.71, respectively, between the FDG PET and MRI-measured lesion volumes, showing that the FDG PET tumor volume is partially contained within the MRI tumor volume. The results showed significant differences in the parameters of SUV in FDG and FMISO PET between patients with and without measurable hypoxic lesions. The quantitative MRI parameters of ADC, T1 max mapping and T2 max mapping were different between hypoxic and normoxic subvolumes. Quantitative MRI, based on free water diffusion and T1 and T2 mapping, seems to be able to identify intra-tumoral hypoxic sub-volumes for additional radiotherapy doses.
Collapse
Affiliation(s)
- Pierrick Gouel
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Françoise Callonnec
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Franchel-Raïs Obongo-Anga
- Department of Surgery, Henri Becquerel Cancer Center and Rouen University Hospital, 76000 Rouen, France
| | - Pierre Bohn
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Emilie Lévêque
- Unit of Clinical Reasearch, Henri Becquerel Cancer Center and Rouen University Hospital, 76000 Rouen, France
| | - David Gensanne
- Department of Radiation Oncology, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108], 76000 Rouen, France
| | - Sébastien Hapdey
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Romain Modzelewski
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Pierre Vera
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
| | - Sébastien Thureau
- Department of Radiology and Nuclear Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, & QuantIF-LITIS [EA (Equipe d'Accueil) 4108-FR CNRS 3638], Faculty of Medicine, University of Rouen, 76000 Rouen, France
- Department of Surgery, Henri Becquerel Cancer Center and Rouen University Hospital, 76000 Rouen, France
| |
Collapse
|
5
|
Gouel P, Decazes P, Vera P, Gardin I, Thureau S, Bohn P. Advances in PET and MRI imaging of tumor hypoxia. Front Med (Lausanne) 2023; 10:1055062. [PMID: 36844199 PMCID: PMC9947663 DOI: 10.3389/fmed.2023.1055062] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Tumor hypoxia is a complex and evolving phenomenon both in time and space. Molecular imaging allows to approach these variations, but the tracers used have their own limitations. PET imaging has the disadvantage of low resolution and must take into account molecular biodistribution, but has the advantage of high targeting accuracy. The relationship between the signal in MRI imaging and oxygen is complex but hopefully it would lead to the detection of truly oxygen-depleted tissue. Different ways of imaging hypoxia are discussed in this review, with nuclear medicine tracers such as [18F]-FMISO, [18F]-FAZA, or [64Cu]-ATSM but also with MRI techniques such as perfusion imaging, diffusion MRI or oxygen-enhanced MRI. Hypoxia is a pejorative factor regarding aggressiveness, tumor dissemination and resistance to treatments. Therefore, having accurate tools is particularly important.
Collapse
Affiliation(s)
- Pierrick Gouel
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Decazes
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Pierre Vera
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Isabelle Gardin
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France
| | - Sébastien Thureau
- QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,Département de Radiothérapie, Centre Henri Becquerel, Rouen, France
| | - Pierre Bohn
- Département d’Imagerie, Centre Henri Becquerel, Rouen, France,QuantIF-LITIS, EA 4108, IRIB, Université de Rouen, Rouen, France,*Correspondence: Pierre Bohn,
| |
Collapse
|
6
|
Tsuchiya H, Matoba M, Nishino Y, Ota K, Doai M, Nagata H, Tuji H. Clinical utility of combined assessments of 4D volumetric perfusion CT, diffusion-weighted MRI and 18F-FDG PET-CT for the prediction of outcomes of head and neck squamous cell carcinoma treated with chemoradiotherapy. Radiat Oncol 2023; 18:24. [PMID: 36747228 PMCID: PMC9901150 DOI: 10.1186/s13014-023-02202-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 01/07/2023] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiparametric imaging has been seen as a route to improved prediction of chemoradiotherapy treatment outcomes. Four-dimensional volumetric perfusion CT (4D PCT) is useful for whole-organ perfusion measurement, as it reflects the heterogeneity of the tumor and its perfusion parameters. However, there has been no study using multiparametric imaging including 4D PCT for the prognostic prediction of chemoradiotherapy. The purpose of this study was to determine whether combining assessments of 4D PCT with diffusion-weighted MRI (DWI) and 18F-fluorodeoxyglucose PET-CT could enhance prognostic accuracy in head and neck squamous cell carcinoma (HNSCC) patients treated with chemoradiotherapy. METHODS We examined 53 patients with HNSCC who underwent 4D PCT, DWI and PET-CT before chemoradiotherapy. The imaging and clinical parameters were assessed the relations to locoregional control (LRC) and progression-free survival (PFS) by logistic regression analyses. A receiver operating characteristic (ROC) analysis was performed to assess the accuracy of the significant parameters identified by the multivariate analysis for the prediction of LRC and PFS. We additionally assessed using the scoring system whether these independent parameters could have a complementary role for the prognostic prediction. RESULTS The median follow-up was 30 months. In multivariate analysis, blood flow (BF; p = 0.02) and blood volume (BV; p = 0.04) were significant prognostic factors for LRC, and BF (p = 0.03) and skewness of the ADC histogram (p = 0.02) were significant prognostic factors for PFS. A significant positive correlation was found between BF and BV (ρ = 0.6, p < 0.001) and between BF and skewness (ρ = 0.46, p < 0.01). The ROC analysis showed that prognostic accuracy for LRC of BF, BV, and combination of BF and BV were 77.8%, 70%, and 92.9%, and that for PFS of BF, skewness, and combination of BF and skewness were 55.6%, 63.2%, and 77.5%, respectively. The scoring system demonstrated that the combination of higher BF and higher BV was significantly associated with better LRC (p = 0.04), and the combination of lower BF and lower skewness was significantly associated with worse PFS (p = 0.004). CONCLUSION A combination of parameters derived from 4DPCT and ADC histograms may enhance prognostic accuracy in HNSCC patients treated with chemoradiotherapy.
Collapse
Affiliation(s)
- Hirokazu Tsuchiya
- grid.411998.c0000 0001 0265 5359Department of Radiology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| | - Munetaka Matoba
- Department of Radiology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa, 920-0293, Japan.
| | - Yuka Nishino
- grid.411998.c0000 0001 0265 5359Department of Radiology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| | - Kiyotaka Ota
- grid.411998.c0000 0001 0265 5359Department of Radiology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| | - Mariko Doai
- grid.411998.c0000 0001 0265 5359Department of Radiology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| | - Hiroji Nagata
- grid.411998.c0000 0001 0265 5359Section of Radiological Technology, Department of Medical Technology, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| | - Hiroyuki Tuji
- grid.411998.c0000 0001 0265 5359Department of Head and Neck Surgery, Kanazawa Medical University, Daigaku 1-1, Uchinada, Kahoku, Ishikawa 920-0293 Japan
| |
Collapse
|
7
|
Goodburn RJ, Philippens MEP, Lefebvre TL, Khalifa A, Bruijnen T, Freedman JN, Waddington DEJ, Younus E, Aliotta E, Meliadò G, Stanescu T, Bano W, Fatemi‐Ardekani A, Wetscherek A, Oelfke U, van den Berg N, Mason RP, van Houdt PJ, Balter JM, Gurney‐Champion OJ. The future of MRI in radiation therapy: Challenges and opportunities for the MR community. Magn Reson Med 2022; 88:2592-2608. [PMID: 36128894 PMCID: PMC9529952 DOI: 10.1002/mrm.29450] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 01/11/2023]
Abstract
Radiation therapy is a major component of cancer treatment pathways worldwide. The main aim of this treatment is to achieve tumor control through the delivery of ionizing radiation while preserving healthy tissues for minimal radiation toxicity. Because radiation therapy relies on accurate localization of the target and surrounding tissues, imaging plays a crucial role throughout the treatment chain. In the treatment planning phase, radiological images are essential for defining target volumes and organs-at-risk, as well as providing elemental composition (e.g., electron density) information for radiation dose calculations. At treatment, onboard imaging informs patient setup and could be used to guide radiation dose placement for sites affected by motion. Imaging is also an important tool for treatment response assessment and treatment plan adaptation. MRI, with its excellent soft tissue contrast and capacity to probe functional tissue properties, holds great untapped potential for transforming treatment paradigms in radiation therapy. The MR in Radiation Therapy ISMRM Study Group was established to provide a forum within the MR community to discuss the unmet needs and fuel opportunities for further advancement of MRI for radiation therapy applications. During the summer of 2021, the study group organized its first virtual workshop, attended by a diverse international group of clinicians, scientists, and clinical physicists, to explore our predictions for the future of MRI in radiation therapy for the next 25 years. This article reviews the main findings from the event and considers the opportunities and challenges of reaching our vision for the future in this expanding field.
Collapse
Affiliation(s)
- Rosie J. Goodburn
- Joint Department of PhysicsInstitute of Cancer Research and Royal Marsden NHS Foundation TrustLondonUnited Kingdom
| | | | - Thierry L. Lefebvre
- Department of PhysicsUniversity of CambridgeCambridgeUnited Kingdom
- Cancer Research UK Cambridge Research InstituteUniversity of CambridgeCambridgeUnited Kingdom
| | - Aly Khalifa
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioCanada
| | - Tom Bruijnen
- Department of RadiotherapyUniversity Medical Center UtrechtUtrechtNetherlands
| | | | - David E. J. Waddington
- Faculty of Medicine and Health, Sydney School of Health Sciences, ACRF Image X InstituteThe University of SydneySydneyNew South WalesAustralia
| | - Eyesha Younus
- Department of Medical Physics, Odette Cancer CentreSunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Eric Aliotta
- Department of Medical PhysicsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Gabriele Meliadò
- Unità Operativa Complessa di Fisica SanitariaAzienda Ospedaliera Universitaria Integrata VeronaVeronaItaly
| | - Teo Stanescu
- Department of Radiation Oncology, University of Toronto and Medical Physics, Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioCanada
| | - Wajiha Bano
- Joint Department of PhysicsInstitute of Cancer Research and Royal Marsden NHS Foundation TrustLondonUnited Kingdom
| | - Ali Fatemi‐Ardekani
- Department of PhysicsJackson State University (JSU)JacksonMississippiUSA
- SpinTecxJacksonMississippiUSA
- Department of Radiation OncologyCommunity Health Systems (CHS) Cancer NetworkJacksonMississippiUSA
| | - Andreas Wetscherek
- Joint Department of PhysicsInstitute of Cancer Research and Royal Marsden NHS Foundation TrustLondonUnited Kingdom
| | - Uwe Oelfke
- Joint Department of PhysicsInstitute of Cancer Research and Royal Marsden NHS Foundation TrustLondonUnited Kingdom
| | - Nico van den Berg
- Department of RadiotherapyUniversity Medical Center UtrechtUtrechtNetherlands
| | - Ralph P. Mason
- Department of RadiologyUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Petra J. van Houdt
- Department of Radiation OncologyNetherlands Cancer InstituteAmsterdamNetherlands
| | - James M. Balter
- Department of Radiation OncologyUniversity of MichiganAnn ArborMichiganUSA
| | - Oliver J. Gurney‐Champion
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam UMCUniversity of AmsterdamAmsterdamNetherlands
| |
Collapse
|
8
|
Mierzwa ML, Aryal M, Lee C, Schipper M, VanTil M, Rivera KM, Swiecicki PL, Casper KA, Malloy KM, Spector ME, Shuman AG, Chinn SB, Prince ME, Stucken CL, Rosko AJ, Lawrence TS, Brenner JC, Rosen B, Schonewolf CA, Shah J, Eisbruch A, Worden FP, Cao Y. Randomized Phase II Study of Physiologic MRI-Directed Adaptive Radiation Boost in Poor Prognosis Head and Neck Cancer. Clin Cancer Res 2022; 28:5049-5057. [PMID: 36107219 PMCID: PMC9773159 DOI: 10.1158/1078-0432.ccr-22-1522] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 07/06/2022] [Accepted: 09/13/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE We conducted a randomized phase II multicenter clinical trial to test the hypothesis that physiologic MRI-based radiotherapy (RT) dose escalation would improve the outcome of patients with poor prognosis head and neck cancer. PATIENTS AND METHODS MRI was acquired at baseline and at RT fraction 10 to create low blood volume/apparent diffusion coefficient maps for RT boost subvolume definition in gross tumor volume. Patients were randomized to receive 70 Gy (standard RT) or 80 Gy to the boost subvolume (RT boost) with concurrent weekly platinum. The primary endpoint was disease-free survival (DFS) with significance defined at a one-sided 0.1 level, and secondary endpoints included locoregional failure (LRF), overall survival (OS), comparison of adverse events and patient reported outcomes (PRO). RESULTS Among 81 randomized patients, neither the primary endpoint of DFS (HR = 0.849, P = 0.31) nor OS (HR = 1.19, P = 0.66) was significantly improved in the RT boost arm. However, the incidence of LRF was significantly improved with the addition of the RT boost (HR = 0.43, P = 0.047). Two-year estimates [90% confidence interval (CI)] of the cumulative incidence of LRF were 40% (27%-53%) in the standard RT arm and 18% (10%-31%) in the RT boost arm. Two-year estimates (90% CI) for DFS were 48% (34%-60%) in the standard RT arm and 57% (43%-69%) in the RT boost arm. There were no significant differences in toxicity or longitudinal differences seen in EORTC QLQ30/HN35 subscales between treatment arms in linear mixed-effects models. CONCLUSIONS Physiologic MRI-based RT boost decreased LRF without a significant increase in grade 3+ toxicity or longitudinal PRO differences, but did not significantly improve DFS or OS. Additional improvements in systemic therapy are likely necessary to realize improvements in DFS and OS.
Collapse
Affiliation(s)
- Michelle L Mierzwa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Madhava Aryal
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Choonik Lee
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Matthew Schipper
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Monica VanTil
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | | | - Paul L. Swiecicki
- Department of Internal Medicine, Medical Oncology, University of Michigan, Ann Arbor, Michigan
| | - Keith A. Casper
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Kelly M. Malloy
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Matthew E. Spector
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Andrew G. Shuman
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Steven B. Chinn
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Mark E.P. Prince
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Chaz L. Stucken
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Andrew J. Rosko
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | | | - J Chad Brenner
- Department of Otolaryngology, University of Michigan, Ann Arbor, Michigan
| | - Benjamin Rosen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | | | - Jennifer Shah
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Avraham Eisbruch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - Francis P. Worden
- Department of Internal Medicine, Medical Oncology, University of Michigan, Ann Arbor, Michigan
| | - Yue Cao
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
9
|
Swartz JE, Smits HJG, Philippens MEP, de Bree R, H A M Kaanders J, Willems SM. Correlation and colocalization of HIF-1α and pimonidazole staining for hypoxia in laryngeal squamous cell carcinomas: A digital, single-cell-based analysis. Oral Oncol 2022; 128:105862. [PMID: 35447566 DOI: 10.1016/j.oraloncology.2022.105862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/31/2022] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Tumor hypoxia results in worse local control and patient survival. We performed a digital, single-cell-based analysis to compare two biomarkers for hypoxia (hypoxia-inducible factor 1-alpha [HIF-1α] and pimonidazole [PIMO]) and their effect on outcome in laryngeal cancer patients treated with accelerated radiotherapy with or without carbogen breathing and nicotinamide (AR versus ARCON). MATERIALS AND METHODS Immunohistochemical staining was performed for HIF-1α and PIMO in consecutive sections of 44 laryngeal cancer patients randomized between AR and ARCON. HIF-1α expression and PIMO-binding were correlated using digital image analysis in QuPath. High-density areas for each biomarker were automatically annotated and staining overlap was analyzed. Kaplan-Meier survival analyses for local control, regional control and disease-free survival were performed to predict a response benefit of ARCON over AR alone for each biomarker. RESULTS 106 Tissue fragments of 44 patients were analyzed. A weak, significant positive correlation was observed between HIF-1α and PIMO positivity on fragment level, but not on patient level. A moderate strength correlation (r = 0.705, p < 0.001) was observed between the number of high-density staining areas for both biomarkers. Staining overlap was poor. HIF-1α expression, PIMO-binding or a combination could not predict a response benefit of ARCON over AR. CONCLUSION Digital image analysis to compare positive cell fractions and staining overlap between two hypoxia biomarkers using open-source software is feasible. Our results highlight that there are distinct differences between HIF-1α and PIMO as hypoxia biomarkers and therefore suggest co-existence of different forms of hypoxia within a single tumor.
Collapse
Affiliation(s)
- Justin E Swartz
- Department of Otorhinolaryngology - Head and Neck Surgery, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Hilde J G Smits
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Remco de Bree
- Department of Head and Neck Surgical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Johannes H A M Kaanders
- Department of Radiation Oncology, University Medical Center Nijmegen, Nijmegen, the Netherlands
| | - Stefan M Willems
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
10
|
Bielak L, Henrik Nicolay N, Ludwig U, Lottner T, Rühle A, Grosu AL, Bock M. Improvement of diffusion weighted MRI by practical B 0 homogenization for head & neck cancer patients undergoing radiation therapy. Phys Med 2022; 97:59-65. [PMID: 35413606 DOI: 10.1016/j.ejmp.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 02/08/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022] Open
Abstract
BACKGROUND MRI is a frequently used tool in radiation therapy planning. For MR-based tumor segmentation, diffusion weighted imaging plays a major role, which can fail due to excessive image artifacts for head and neck cancer imaging. Here, an easy-to-use setup is presented for imaging of head and neck cancer patients in a radiotherapy thermoplastic fixation mask. METHODS In a prospective head and neck cancer study, MRI data of 29 patients has been acquired at 3 different time points during radiation treatment. The data was analyzed with respect to Nyquist ghosting artifacts in the diffusion images in conventional single shot and readout segmented EPI sequences. For 9 patients, an improved setup with water bags for B0 homogenization was used, and the impact on artifact frequency was analyzed. Additionally, volunteer measurements with B0 fieldmaps are presented. RESULTS The placement of water bags to the sides of the head during MRI measurements significantly reduces artefacts in diffusion MRI. The number of artifact-free images in readout segmented EPI increased from 74% to 95% of the cases. Volunteer measurements showed a significant increase in B0 homogeneity across slices (head foot direction) as well as within each slice. CONCLUSIONS The placement of water bags for B0 homogenization is easy to implement, cost-efficient and does not impact patient comfort. Therefore, if very sophisticated soft- or hardware solutions are not present at a given site, or cannot be implemented due to restrictions from the thermoplastic mask, this is an excellent alternative to reduce artifacts in diffusion weighted imaging.
Collapse
Affiliation(s)
- Lars Bielak
- Dept. 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.
| | - Nils Henrik Nicolay
- Dept. 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
| | - Ute Ludwig
- Dept. of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Lottner
- Dept. of Radiology, Medical Physics, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Rühle
- Dept. of Radiation Oncology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anca-Ligia Grosu
- Dept. 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
| | - Michael Bock
- Dept. 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
| |
Collapse
|
11
|
Welz S, Paulsen F, Pfannenberg C, Reimold M, Reischl G, Nikolaou K, La Fougère C, Alber M, Belka C, Zips D, Thorwarth D. Dose escalation to hypoxic subvolumes in head and neck cancer: A randomized phase II study using dynamic [ 18F]FMISO PET/CT. Radiother Oncol 2022; 171:30-36. [PMID: 35395276 DOI: 10.1016/j.radonc.2022.03.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND PURPOSE Tumor hypoxia is a major cause of resistance to radiochemotherapy in locally advanced head-and-neck cancer (LASCCHN). We present results of a randomized phase II trial on hypoxia dose escalation (DE) in LASCCHN based on dynamic [18F]FMISO (dynFMISO) positron emission tomography (PET). The purpose was to confirm the prognostic value of hypoxia PET and assess feasibility, toxicity and efficacy of hypoxia-DE. MATERIALS AND METHODS Patients with LASCCHN underwent baseline dynFMISO PET/CT. Hypoxic volumes (HV) were derived from dynFMISO data. Patients with hypoxic tumors (HV>0) were randomized into standard radiotherapy (ST: 70Gy/35fx) or dose escalation (DE: 77Gy/35fx) to the HV. Patients with non-hypoxic tumors were treated with ST. After a minimum follow-up of 2 years, feasibility, acute/late toxicity and local control (LC) were analyzed. RESULTS The study was closed prematurely due to slow accrual. Between 2009 and 2017, 53 patients were enrolled, 39 (74%) had hypoxic tumors and were randomized into ST or DE. For non-hypoxic patients, 100% 5-year LC was observed compared to 74% in patients with hypoxic tumors (p=0.039). The difference in 5-year LC between DE (16/19) and ST (10/17) was 25%, p=0.150. No relevant differences related to acute and late toxicities between the groups were observed. CONCLUSION This study confirmed the prognostic value of hypoxia PET in LASCCHN for LC. Outcome after hypoxia DE appears promising and may support the concept of DE. Slow accrual and premature closure may partly be due to a high complexity of the study setup which needs to be considered for future multicenter trials.
Collapse
Affiliation(s)
- Stefan Welz
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Frank Paulsen
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christina Pfannenberg
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Matthias Reimold
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Gerald Reischl
- Department of Preclinical Imaging and Radiopharmacy, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) "Image Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Germany
| | - Konstantin Nikolaou
- Department of Radiology, Diagnostic and Interventional Radiology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Christian La Fougère
- Department of Nuclear Medicine, University Hospital Tübingen, University of Tübingen, Tübingen, Germany
| | - Markus Alber
- Section for Medical Physics, Department of Radiation Oncology, Heidelberg University, Heidelberg, Germany
| | - Claus Belka
- Department of Radiation Oncology, University of Munich, Germany; Department of Radiation Oncology, LMU Munich, Germany
| | - Daniel Zips
- Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), partner site Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany; Section for Biomedical Physics, Department of Radiation Oncology, University Hospital Tübingen, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
12
|
Liu J, Lu J, Li W. Transcriptome analysis reveals the prognostic and immune infiltration characteristics of glycolysis and hypoxia in head and neck squamous cell carcinoma. BMC Cancer 2022; 22:352. [PMID: 35361159 PMCID: PMC8969218 DOI: 10.1186/s12885-022-09449-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND This study aims to construct a new prognostic gene signature in survival prediction and risk stratification for patients with Head and neck squamous cell carcinoma (HNSCC). METHOD The transcriptome profiling data and hallmark gene sets in the Molecular Signatures Database was used to explore the cancer hallmarks most relevant to the prognosis of HNSCC patients. Differential gene expression analysis, weighted gene co-expression network analysis, univariate COX regression analysis, random forest algorithm and multiple combinatorial screening were used to construct the prognostic gene signature. The predictive ability of gene signature was verified in the TCGA HNSCC cohort as the training set and the GEO HNSCC cohorts (GSE41613 and GSE42743) as the validation sets, respectively. Moreover, the correlations between risk scores and immune infiltration patterns, as well as risk scores and genomic changes were explored. RESULTS A total of 3391 differentially expressed genes in HNSCC were screened. Glycolysis and hypoxia were screened as the main risk factors for OS in HNSCC. Using univariate Cox analysis, 97 prognostic candidates were identified (P < 0.05). Top 10 important genes were then screened out by random forest. Using multiple combinatorial screening, a combination with less genes and more significant P value was used to construct the prognostic gene signature (RNF144A, STC1, P4HA1, FMNL3, ANO1, BASP1, MME, PLEKHG2 and DKK1). Kaplan-Meier analysis showed that patients with higher risk scores had worse overall survival (p < 0.001). The ROC curve showed that the risk score had a good predictive efficiency (AUC > 0.66). Subsequently, the predictive ability of the risk score was verified in the validation sets. Moreover, the two-factor survival analysis combining the cancer hallmarks and risk scores suggested that HNSCC patients with the high hypoxia or glycolysis & high risk-score showed the worst prognosis. Besides, a nomogram based on the nine-gene signature was established for clinical practice. Furthermore, the risk score was significantly related to tumor immune infiltration profiles and genome changes. CONCLUSION This nine-gene signature associated with glycolysis and hypoxia can not only be used for prognosis prediction and risk stratification, but also may be a potential therapeutic target for patients with HNSCC.
Collapse
Affiliation(s)
- Jun Liu
- Reproductive Medicine Center, Yue Bei People's Hospital, Shantou University Medical College, 133 Huimin South Road, Shaoguan, 512025, China
- Medical Research Center, Yue Bei People's Hospital, Shantou University Medical College, Shaoguan, 512025, China
| | - Jianjun Lu
- Department of Medical Affairs, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510080, China
| | - Wenli Li
- Reproductive Medicine Center, Yue Bei People's Hospital, Shantou University Medical College, 133 Huimin South Road, Shaoguan, 512025, China
| |
Collapse
|
13
|
Lefebvre TL, Brown E, Hacker L, Else T, Oraiopoulou ME, Tomaszewski MR, Jena R, Bohndiek SE. The Potential of Photoacoustic Imaging in Radiation Oncology. Front Oncol 2022; 12:803777. [PMID: 35311156 PMCID: PMC8928467 DOI: 10.3389/fonc.2022.803777] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
Radiotherapy is recognized globally as a mainstay of treatment in most solid tumors and is essential in both curative and palliative settings. Ionizing radiation is frequently combined with surgery, either preoperatively or postoperatively, and with systemic chemotherapy. Recent advances in imaging have enabled precise targeting of solid lesions yet substantial intratumoral heterogeneity means that treatment planning and monitoring remains a clinical challenge as therapy response can take weeks to manifest on conventional imaging and early indications of progression can be misleading. Photoacoustic imaging (PAI) is an emerging modality for molecular imaging of cancer, enabling non-invasive assessment of endogenous tissue chromophores with optical contrast at unprecedented spatio-temporal resolution. Preclinical studies in mouse models have shown that PAI could be used to assess response to radiotherapy and chemoradiotherapy based on changes in the tumor vascular architecture and blood oxygen saturation, which are closely linked to tumor hypoxia. Given the strong relationship between hypoxia and radio-resistance, PAI assessment of the tumor microenvironment has the potential to be applied longitudinally during radiotherapy to detect resistance at much earlier time-points than currently achieved by size measurements and tailor treatments based on tumor oxygen availability and vascular heterogeneity. Here, we review the current state-of-the-art in PAI in the context of radiotherapy research. Based on these studies, we identify promising applications of PAI in radiation oncology and discuss the future potential and outstanding challenges in the development of translational PAI biomarkers of early response to radiotherapy.
Collapse
Affiliation(s)
- Thierry L. Lefebvre
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Emma Brown
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Lina Hacker
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Else
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Mariam-Eleni Oraiopoulou
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Michal R. Tomaszewski
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Rajesh Jena
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Sarah E. Bohndiek
- Department of Physics, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
14
|
The value of plasma hypoxia markers for predicting imaging-based hypoxia in patients with head-and-neck cancers undergoing definitive chemoradiation. Clin Transl Radiat Oncol 2022; 33:120-127. [PMID: 35243023 PMCID: PMC8881198 DOI: 10.1016/j.ctro.2022.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 11/22/2022] Open
Abstract
Higher osteopontin plasma levels correlate with more hypoxic tumors at baseline. Increased baseline osteopontin levels are associated with residual tumor hypoxia. Absent early hypoxia response is linked with higher VEGF and CTGF levels in week 5. Plasma hypoxic markers may serve as biomarkers favoring radiotherapy personalization.
Background Methods Results Conclusion
Collapse
|
15
|
Rühle A, Wiedenmann N, Fennell JT, Mix M, Ruf J, Stoian R, Thomsen AR, Vaupel P, Baltas D, Grosu AL, Nicolay NH. Interleukin-6 as surrogate marker for imaging-based hypoxia dynamics in patients with head-and-neck cancers undergoing definitive chemoradiation-results from a prospective pilot trial. Eur J Nucl Med Mol Imaging 2021; 49:1650-1660. [PMID: 34773163 PMCID: PMC8940848 DOI: 10.1007/s00259-021-05602-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022]
Abstract
Purpose Intratumoral hypoxia increases resistance of head-and-neck squamous cell carcinoma (HNSCC) to radiotherapy. [18F]FMISO PET imaging enables noninvasive hypoxia monitoring, though requiring complex logistical efforts. We investigated the role of plasma interleukin-6 (IL-6) as potential surrogate parameter for intratumoral hypoxia in HNSCC using [18F]FMISO PET/CT as reference. Methods Within a prospective trial, serial blood samples of 27 HNSCC patients undergoing definitive chemoradiation were collected to analyze plasma IL-6 levels. Intratumoral hypoxia was assessed in treatment weeks 0, 2, and 5 using [18F]FMISO PET/CT imaging. The association between PET-based hypoxia and IL-6 was examined using Pearson’s correlation and multiple regression analyses, and the diagnostic power of IL-6 for tumor hypoxia response prediction was determined with receiver-operating characteristic analyses. Results Mean IL-6 concentrations were 15.1, 19.6, and 31.0 pg/mL at baseline, week 2 and week 5, respectively. Smoking (p=0.050) and reduced performance status (p=0.011) resulted in higher IL-6 levels, whereas tumor (p=0.427) and nodal stages (p=0.334), tumor localization (p=0.439), and HPV status (p=0.294) had no influence. IL-6 levels strongly correlated with the intratumoral hypoxic subvolume during treatment (baseline: r=0.775, p<0.001; week 2: r=0.553, p=0.007; week 5: r=0.734, p<0.001). IL-6 levels in week 2 were higher in patients with absent early tumor hypoxia response (p=0.016) and predicted early hypoxia response (AUC=0.822, p=0.031). Increased IL-6 levels at week 5 resulted in a trend towards reduced progression-free survival (p=0.078) and overall survival (p=0.013). Conclusion Plasma IL-6 is a promising surrogate marker for tumor hypoxia dynamics in HNSCC patients and may facilitate hypoxia-directed personalized radiotherapy concepts. Trial registration The prospective trial was registered in the German Clinical Trial Register (DRKS00003830). Registered 20 August 2015 Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05602-x.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jamina T Fennell
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Raluca Stoian
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas R Thomsen
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Peter Vaupel
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Germany. .,German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
16
|
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
|
17
|
Connor S, Sit C, Anjari M, Szyszko T, Dunn J, Pai I, Cook G, Goh V. Correlations between DW-MRI and 18 F-FDG PET/CT parameters in head and neck squamous cell carcinoma following definitive chemo-radiotherapy. Cancer Rep (Hoboken) 2021; 4:e1360. [PMID: 33960739 PMCID: PMC8388179 DOI: 10.1002/cnr2.1360] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Posttreatment diffusion-weighted magnetic resonance imaging (DW-MRI) and 18F-fluorodeoxygluocose (18 F-FDG) positron emission tomography (PET) with computed tomography (PET/CT) have potential prognostic value following chemo-radiotherapy (CRT) for head and neck squamous cell carcinoma (HNSCC). Correlations between these PET/CT (standardized uptake value or SUV) and DW-MRI (apparent diffusion coefficient or ADC) parameters have only been previously explored in the pretreatment setting. AIM To evaluate stage III and IV HNSCC at 12-weeks post-CRT for the correlation between SUVmax and ADC values and their interval changes from pretreatment imaging. METHODS Fifty-six patients (45 male, 11 female, mean age 59.9 + - 7.38) with stage 3 and 4 HNSCC patients underwent 12-week posttreatment DW-MRI and 18 F-FDG PET/CT studies in this prospective study. There were 41/56 patients in the cohort with human papilloma virus-related oropharyngeal cancer (HPV OPC). DW-MRI (ADCmax and ADCmin) and 18 F-FDG PET/CT (SUVmax and SUVmax ratio to liver) parameters were measured at the site of primary tumors (n = 48) and the largest lymph nodes (n = 52). Kendall's tau evaluated the correlation between DW-MRI and 18 F-FDG PET/CT parameters. Mann-Whitney test compared the post-CRT PET/CT and DW-MRI parameters between those participants with and without 2-year disease-free survival (DFS). RESULTS There was no correlation between DW-MRI and 18 F-FDG PET/CT parameters on 12-week posttreatment imaging (P = .455-.794; tau = -0.075-0.25) or their interval changes from pretreatment to 12-week posttreatment imaging (P = .1-.946; tau = -0.194-0.044). The primary tumor ADCmean (P = .03) and the interval change in nodal ADCmin (P = .05) predicted 2-year DFS but none of the 18 F-FDG PET/CT parameters were associated with 2-year DFS. CONCLUSIONS There is no correlation between the quantitative DWI-MRI and 18 F-FDG PET/CT parameters derived from 12-week post-CRT studies. These parameters may be independent biomarkers however in this HPV OPC dominant cohort, only selected ADC parameters demonstrated prognostic significance. Study was prospectively registered at http://www.controlled-trials.com/ISRCTN58327080.
Collapse
Affiliation(s)
- Steve Connor
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Hospital, King's CollegeLondonUK
- Department of NeuroradiologyKing's College Hospital NHS Foundation TrustLondonUK
- Department of RadiologyGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Cherry Sit
- Department of RadiologyGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Mustafa Anjari
- Department of RadiologyGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Teresa Szyszko
- King's College London & Guy's and St. Thomas' PET CentreLondonUK
| | - Joel Dunn
- King's College London & Guy's and St. Thomas' PET CentreLondonUK
| | - Irumee Pai
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Hospital, King's CollegeLondonUK
- Department of OtolaryngologyGuy's and St Thomas' NHS Foundation TrustLondonUK
| | - Gary Cook
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Hospital, King's CollegeLondonUK
- King's College London & Guy's and St. Thomas' PET CentreLondonUK
| | - Vicky Goh
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Hospital, King's CollegeLondonUK
- Department of RadiologyGuy's and St Thomas' NHS Foundation TrustLondonUK
| |
Collapse
|
18
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Strahlenther Onkol 2021; 197:1-23. [PMID: 34259912 DOI: 10.1007/s00066-021-01812-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Christoph Henkenberens
- Department of Radiotherapy and Special Oncology, Medical School Hannover, Hannover, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany.
| |
Collapse
|
19
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Nuklearmedizin 2021; 60:326-343. [PMID: 34261141 DOI: 10.1055/a-1525-7029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | | | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiotherapy and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | | |
Collapse
|
20
|
Rühle A, Nicolay NH. [Aggressive radiotherapy de-escalation for HPV-associated oropharyngeal carcinoma based on hypoxia dynamics]. Strahlenther Onkol 2021; 197:570-573. [PMID: 33765185 PMCID: PMC8154749 DOI: 10.1007/s00066-021-01765-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2021] [Indexed: 01/21/2023]
Affiliation(s)
- Alexander Rühle
- Klinik für Strahlenheilkunde, Universitätsklinikum Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnerstandort Freiburg, Deutsches Krebsforschungszentrum (dkfz), Heidelberg, Deutschland.
| | - Nils H Nicolay
- Klinik für Strahlenheilkunde, Universitätsklinikum Freiburg, Robert-Koch-Str. 3, 79106, Freiburg, Deutschland.
- Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnerstandort Freiburg, Deutsches Krebsforschungszentrum (dkfz), Heidelberg, Deutschland.
| |
Collapse
|
21
|
Rühle A, Grosu AL, Nicolay NH. De-Escalation Strategies of (Chemo)Radiation for Head-and-Neck Squamous Cell Cancers-HPV and Beyond. Cancers (Basel) 2021; 13:2204. [PMID: 34064321 PMCID: PMC8124930 DOI: 10.3390/cancers13092204] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/29/2021] [Accepted: 05/01/2021] [Indexed: 12/17/2022] Open
Abstract
Oncological outcomes for head-and-neck squamous cell carcinoma (HNSCC) patients are still unsatisfactory, especially for advanced tumor stages. Besides the moderate survival rates, the prevalence of severe treatment-induced normal tissue toxicities is high after multimodal cancer treatments, both causing significant morbidity and decreasing quality of life of surviving patients. Therefore, risk-adapted and individualized treatment approaches are urgently needed for HNSCC patients to optimize the therapeutic gain. It has been a well-known fact that especially HPV-positive oropharyngeal squamous cell carcinoma (OSCC) patients exhibit an excellent prognosis and may therefore be subject to overtreatment, resulting in long-term treatment-related toxicities. Regarding the superior prognosis of HPV-positive OSCC patients, treatment de-escalation strategies are currently investigated in several clinical trials, and HPV-positive OSCC may potentially serve as a model for treatment de-escalation also for other types of HNSCC. We performed a literature search for both published and ongoing clinical trials and critically discussed the presented concepts and results. Radiotherapy dose or volume reduction, omission or modification of concomitant chemotherapy, and usage of induction chemotherapy are common treatment de-escalation strategies that are pursued in clinical trials for biologically selected subgroups of HNSCC patients. While promising data have been reported from various Phase II trials, evidence from Phase III de-escalation trials is either lacking or has failed to demonstrate comparable outcomes for de-escalated treatments. Therefore, further data and a refinement of biological HNSCC stratification are required before deescalated radiation treatments can be recommended outside of clinical trials.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, University of Freiburg—Medical Center, Robert-Koch-Str. 3, 79106 Freiburg, Germany; (A.R.); (A.-L.G.)
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University of Freiburg—Medical Center, Robert-Koch-Str. 3, 79106 Freiburg, Germany; (A.R.); (A.-L.G.)
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nils H. Nicolay
- Department of Radiation Oncology, University of Freiburg—Medical Center, Robert-Koch-Str. 3, 79106 Freiburg, Germany; (A.R.); (A.-L.G.)
- German Cancer Consortium (DKTK) Partner Site Freiburg, German Cancer Research Center (dkfz), Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
22
|
Zeng F, Zhang Y, Han X, Zeng M, Gao Y, Weng J. Employing hypoxia characterization to predict tumour immune microenvironment, treatment sensitivity and prognosis in hepatocellular carcinoma. Comput Struct Biotechnol J 2021; 19:2775-2789. [PMID: 34093992 PMCID: PMC8134035 DOI: 10.1016/j.csbj.2021.03.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
The hypoxic microenvironment was recognized as a major driving force of the malignant phenotype in hepatocellular carcinoma (HCC), which contributes to tumour immune microenvironment (TIM) remodeling and tumor progression. Dysregulated hypoxia-related genes (HRGs) result in treatment resistance and poor prognosis by reshaping tumor cellular activities and metabolism. Approaches to identify the relationship between hypoxia and tumor progression provided new sight for improving tumor treatment and prognosis. But, few practical tools, forecasting relationship between hypoxia, TIM, treatment sensitivity and prognosis in HCC were reported. Here, we pooled mRNA transcriptome and clinical pathology data from the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), and later developed a hypoxia risk model including four HRGs (DCN, DDIT4, PRKCA and NDRG1). The high-risk group displayed poor clinical characteristics, a malignant phenotype with carcinogenesis/proliferation pathways activation (MTORC1 and E2F) and immunosuppressive TIM (decreased immune cell infiltrations and upregulated immunosuppressive cytokines). Meanwhile, activated B cells, effector memory CD8 T cells and EZH2 deregulation were associated with patient’s survival, which might be the core changes of HCC hypoxia. Finally, we validated the ability of the hypoxia risk model to predict treatment sensitivity and found high hypoxia risk patients had poor responses to HCC treatment, including surgical resection, Sorafenib, Transarterial Chemoembolization (TACE) and immunotherapy. In conclusion, based on 4 HRGs, we developed and validated a hypoxia risk model to reflect pathological features, evaluate TIM landscape, predict treatment sensitivity and compounds specific to hypoxia signatures in HCC patients.
Collapse
Affiliation(s)
- Fanhong Zeng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Xu Han
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Min Zeng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Yi Gao
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| | - Jun Weng
- Department of Hepatobiliary Surgery II, Guangdong Provincial Research Center for Artificial Organ and Tissue Engineering, Guangzhou Clinical Research and Transformation Center for Artificial Liver, Institute of Regenerative Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Nehmeh SA, Moussa MB, Lee N, Zanzonico P, Gönen M, Humm JL, Schöder H. Comparison of FDG and FMISO uptakes and distributions in head and neck squamous cell cancer tumors. EJNMMI Res 2021; 11:38. [PMID: 33855685 PMCID: PMC8046891 DOI: 10.1186/s13550-021-00767-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/26/2021] [Indexed: 12/02/2022] Open
Abstract
Purpose Glycolysis is increased by hypoxia, suggesting a possible correlation between the accumulation of 2-[18F]fluoro-2-deoxy-D-glucose (FDG) in malignant tumors and regional hypoxia defined by 1H-1-(3-[18F]fluoro-2-hydroxypropyl)-2-nitroimidazole (FMISO) PET. The aim of this study is to investigate the intra-tumoral spatial distribution and quantitative relationship between FDG and FMISO in a cohort of head and neck squamous cell cancer (HNSCC) patients. Methods Twenty HNSCC patients with 20 primary tumors and 19 metastatic lymph nodes (LNs) underwent FDG and FMISO PET within 1 week. The metabolic target volume (MTV) was defined on the FDG PET images using a region growing algorithm. The hypoxic volume (HV) was defined by the volume of voxels in an FMISO image within the MTV that satisfy a tumor-to-blood ratio (T/B) greater than 1.2. FDG and FMISO lesions were co-registered, and a voxel-by-voxel correlation between the two datasets was performed. FDG and FMISO TVs’ SUVs were also compared as well as the intra-tumoral homogeneity of the two radiotracers. Separate analysis was performed for the primary tumors and LNs. Results Twenty-six percent of the primary tumors and 15% of LNs showed a strong correlation (R > 0.7) between FDG and FMISO intra-tumor distributions when considering the MTV. For the HV, only 19% of primary tumors and 12% of LN were strongly correlated. A weak and moderate correlation existed between the two markers SUVavg, and SUVmax in the case of the primary tumors, respectively. However, this was not the case for the LNs. Good concordances were also observed between the primary tumor’s and LNs HV SUVavgs as well as between the corresponding hypoxic fractions (HF’s). Conclusions A moderate correlation between FDG and hypoxia radiotracer distribution, as measured by FMISO, seems to exist for primary tumors. However, discordant results were found in the case of LNs. Hypoxia appears to be the dominant driver of high FDG uptake in selected tumors only, and therefore FDG PET images cannot be used as a universal surrogate to identify or predict intra-tumor hypoxia.
Collapse
Affiliation(s)
- Sadek A Nehmeh
- Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA. .,Weill Cornell Medical College, New York, NY, 10021, USA.
| | - Mohamed B Moussa
- Chemistry Department, Stony Brook University, Stony Brook, NY, USA
| | - Nancy Lee
- Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Pat Zanzonico
- Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - John L Humm
- Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Heiko Schöder
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
24
|
Boeke S, Mönnich D, van Timmeren JE, Balermpas P. MR-Guided Radiotherapy for Head and Neck Cancer: Current Developments, Perspectives, and Challenges. Front Oncol 2021; 11:616156. [PMID: 33816247 PMCID: PMC8017313 DOI: 10.3389/fonc.2021.616156] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
Based on the development of new hybrid machines consisting of an MRI and a linear accelerator, magnetic resonance image guided radiotherapy (MRgRT) has revolutionized the field of adaptive treatment in recent years. Although an increasing number of studies have been published, investigating technical and clinical aspects of this technique for various indications, utilizations of MRgRT for adaptive treatment of head and neck cancer (HNC) remains in its infancy. Yet, the possible benefits of this novel technology for HNC patients, allowing for better soft-tissue delineation, intra- and interfractional treatment monitoring and more frequent plan adaptations appear more than obvious. At the same time, new technical, clinical, and logistic challenges emerge. The purpose of this article is to summarize and discuss the rationale, recent developments, and future perspectives of this promising radiotherapy modality for treating HNC.
Collapse
Affiliation(s)
- Simon Boeke
- Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - David Mönnich
- Section for Biomedical Physics, Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | - Panagiotis Balermpas
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
25
|
Rühle A, Grosu AL, Wiedenmann N, Stoian R, Haehl E, Zamboglou C, Baltas D, Werner M, Kayser G, Nicolay NH. Immunohistochemistry-based hypoxia-immune prognostic classifier for head-and-neck cancer patients undergoing chemoradiation - Post-hoc analysis from a prospective imaging trial. Radiother Oncol 2021; 159:75-81. [PMID: 33753155 DOI: 10.1016/j.radonc.2021.03.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE As both tumor hypoxia and an immunosuppressing tumor microenvironment hamper the anti-tumor activity of radiotherapy in head-and-neck squamous cell carcinoma (HNSCC), we aimed to develop an immunohistochemistry-based hypoxia-immune classifier. METHODS 39 patients receiving definitive chemoradiation for HNSCC within a prospective trial were included in this analysis. Baseline tumor samples were analyzed for the hypoxia marker carbonic anhydrase IX (CAIX) and tumor-infiltrating lymphocytes (TILs) and were correlated with [18F]-misonidazole ([18F]FMISO) PET measurements. The impact of the biomarkers on the locoregional control (LRC) was examined using Cox analyses and concordance index statistics. RESULTS Low CAIX (HR = 0.352, 95%CI 0.124-1.001, p = 0.050) and high TIL levels (HR = 0.308, 95%CI 0.114-0.828, p = 0.020) were independent parameters for improved LRC and did not correlate with each other (Spearman's ρ = 0.034, p = 0.846). Harrell's C was 0.66 for CAIX and TIL levels alone and 0.71 for the combination. 2-year LRC was 73%, 62% and 11% for the prognostically good (CAIXlow/TILhigh), intermediate (CAIXlow/TILlow or CAIXhigh/TILhigh) and poor groups (CAIXhigh/TILlow), respectively (p = 0.001). Focusing on T lymphocytes, the hypoxia-immune classifier could still stratify between favorable (CAIXlow/CD3 + TILhigh), intermediate (CAIXlow/CD3 + TILlow or CAIXhigh/CD3 + TILhigh) and poor subgroups (CAIXhigh/CD3 + TILlow) with a 2-year LRC of 80%, 59% and 14%, respectively (p = 0.001). There was a positive correlation between baseline CAIX levels and [18F]FMISO SUV in week 2 of chemoradiation (ρ = 0.324, p = 0.050), indicating an association between higher baseline CAIX expression and tumor hypoxia persistence. CONCLUSION We developed a clinically feasible hypoxia-immune prognostic classifier for HNSCC patients based on pre-treatment immunohistochemistry. However, external validation is required to determine the prognostic value and the potential usage for personalized radiation oncology.
Collapse
Affiliation(s)
- Alexander Rühle
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raluca Stoian
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Erik Haehl
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dimos Baltas
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Werner
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Surgical Pathology, Department of Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gian Kayser
- German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany; Institute of Surgical Pathology, Department of Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
26
|
Mayer P, Kraft A, Witzel HR, Marnet N, Hörner N, Roth W, Heinrich S, Hackert T, Bergmann F, Kauczor HU, Klauss M, Gaida MM. Restricted Water Diffusion in Diffusion-Weighted Magnetic Resonance Imaging in Pancreatic Cancer is Associated with Tumor Hypoxia. Cancers (Basel) 2020; 13:cancers13010089. [PMID: 33396818 PMCID: PMC7801953 DOI: 10.3390/cancers13010089] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Pancreatic cancer is characterized by a dense network of connective tissue surrounding clusters of cancer cells, the so-called stroma. This ubiquitous connective tissue impairs the delivery of oxygen to cancer cells. This results in hypoxia, which renders the cancer more aggressive and more resistant to treatment. In the present study, we investigated whether the extent of hypoxia in pancreatic cancer can be predicted by magnetic resonance imaging (MRI), a widely used medical imaging technique. More specifically, we used an MRI sequence which can quantitate the random motion (i.e., diffusion) of water molecules within the cancer tissue, namely diffusion-weighted (DW) MRI. We found that the random motion of water molecules is lower in cancer lesions with high hypoxia compared to those with low hypoxia. The findings from our study imply that DW-MRI can be used to identify pancreatic cancer lesions with high hypoxia which are at high risk for treatment failure. Abstract Hypoxia is a hallmark of pancreatic cancer (PDAC) due to its compact and extensive fibrotic tumor stroma. Hypoxia contributes to high lethality of this disease, by inducing a more malignant phenotype and resistance to radiation and chemotherapy. Thus, non-invasive methods to quantify hypoxia could be helpful for treatment decisions, for monitoring, especially in non-resectable tumors, or to optimize personalized therapy. In the present study, we investigated whether tumor hypoxia in PDAC is reflected by diffusion-weighted magnetic resonance imaging (DW-MRI), a functional imaging technique, frequently used in clinical practice for identification and characterization of pancreatic lesions. DW-MRI assesses the tissue microarchitecture by measuring the diffusion of water molecules, which is more restricted in highly compact tissues. As reliable surrogate markers for hypoxia, we determined Blimp-1 (B-lymphocyte induced maturation protein), a transcription factor, as well as vascular endothelial growth factor (VEGF), which are up-regulated in response to hypoxia. In 42 PDAC patients, we observed a close association between restricted water diffusion in DW-MRI and tumor hypoxia in matched samples, as expressed by high levels of Blimp-1 and VEGF in tissue samples of the respective patients. In summary, our data show that DW-MRI is well suited for the evaluation of tumor hypoxia in PDAC and could potentially be used for the identification of lesions with a high hypoxic fraction, which are at high risk for failure of radiochemotherapy.
Collapse
Affiliation(s)
- Philipp Mayer
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
- Correspondence: ; Tel.: +49-6221-5637-345
| | - Anne Kraft
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Hagen R. Witzel
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nicole Marnet
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Nina Hörner
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Wilfried Roth
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
| | - Stefan Heinrich
- Department of Surgery, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany;
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany;
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany;
- Clinical Pathology, Klinikum Darmstadt GmbH, 64283 Darmstadt, Germany
| | - Hans-Ulrich Kauczor
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Miriam Klauss
- Clinic for Diagnostic and Interventional Radiology, University Hospital Heidelberg, 69120 Heidelberg, Germany; (H.-U.K.); (M.K.)
| | - Matthias M. Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany; (A.K.); (H.R.W.); (N.M.); (N.H.); (W.R.); (M.M.G.)
- Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, 55131 Mainz, Germany
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, JGU-Mainz and TRON, Translational Oncology at the University Medical Center, JGU-Mainz, 55131 Mainz, Germany
| |
Collapse
|
27
|
Nicolay NH, Rühle A, Wiedenmann N, Niedermann G, Mix M, Weber WA, Baltas D, Werner M, Kayser G, Grosu AL. Lymphocyte Infiltration Determines the Hypoxia-Dependent Response to Definitive Chemoradiation in Head-and-Neck Cancer: Results from a Prospective Imaging Trial. J Nucl Med 2020; 62:471-478. [PMID: 32859699 DOI: 10.2967/jnumed.120.248633] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/28/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor hypoxia in head-and-neck squamous cell carcinoma (HNSCC) leads to an immunosuppressive microenvironment and reduces the response to radiotherapy. In this prospective imaging trial, we investigated potential interactions between functional hypoxia imaging and infiltrating lymphocyte levels as a potential predictor for treatment response in HNSCC patients. Methods: In total, 49 patients receiving definitive chemoradiation for locally advanced HNSCCs underwent pretherapeutic biopsies and peritherapeutic hypoxia imaging using 18F-misonidazole PET at weeks 0, 2, and 5 during chemoradiation. Hematoxylin-eosin and immunohistochemical stainings for tumor-infiltrating lymphocytes, tissue-based hypoxia, and microvascular markers were analyzed and correlated with the longitudinal hypoxia dynamics and patient outcomes. Results: High levels of tumor-infiltrating total lymphocytes correlated with superior locoregional control (LRC) (hazard ratio [HR], 0.279; P = 0.011) and progression-free survival (PFS) (HR, 0.276; P = 0.006). Similarly, early resolution of 18F-misonidazole PET-detected tumor hypoxia quantified by 18F-misonidazole dynamics between weeks 0 and 2 of chemoradiation was associated with improved LRC (HR, 0.321; P = 0.015) and PFS (HR, 0.402; P = 0.043). Outcomes in the favorable early hypoxia resolution subgroup significantly depended on infiltrating lymphocyte counts, with patients who showed both an early hypoxia response and high lymphocyte infiltration levels exhibiting significantly improved LRC (HR, 0.259; P = 0.036) and PFS (HR, 0.242; P = 0.017) compared with patients with an early hypoxia response but low lymphocyte counts. These patients exhibited oncologic results comparable to those of patients with no hypoxia response within the first 2 wk of chemoradiation. Conclusion: This analysis established a clinical hypoxia-immune score that predicted treatment responses and outcomes in HNSCC patients undergoing chemoradiation and may help to devise novel concepts for biology-driven personalization of chemoradiation.
Collapse
Affiliation(s)
- Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany .,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Alexander Rühle
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Nicole Wiedenmann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Gabriele Niedermann
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany; and
| | - Dimos Baltas
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| | - Martin Werner
- German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Gian Kayser
- German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany.,Institute of Surgical Pathology, Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Partner Site Freiburg and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
28
|
Rühle A, Grosu AL, Wiedenmann N, Mix M, Stoian R, Niedermann G, Baltas D, Werner M, Weber WA, Kayser G, Nicolay NH. Hypoxia dynamics on FMISO-PET in combination with PD-1/PD-L1 expression has an impact on the clinical outcome of patients with Head-and-neck Squamous Cell Carcinoma undergoing Chemoradiation. Am J Cancer Res 2020; 10:9395-9406. [PMID: 32802199 PMCID: PMC7415814 DOI: 10.7150/thno.48392] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated hypoxia influences the radiation response of head-and-neck cancer (HNSCC) patients, and a lack of early hypoxia resolution during treatment considerably deteriorates outcomes. As the detrimental effects of hypoxia are partly related to the induction of an immunosuppressive microenvironment, we investigated the interaction between tumor hypoxia dynamics and the PD-1/PD-L1 axis in HNSCC patients undergoing chemoradiation and its relevance for patient outcomes in a prospective trial. Methods: 49 patients treated with definitive chemoradiation for locally advanced HNSCC were enrolled in this trial and received longitudinal hypoxia PET imaging using fluorine-18 misonidazole ([18F]FMISO) at weeks 0, 2 and 5 during treatment. Pre-therapeutic tumor biopsies were immunohistochemically analyzed regarding the PD-1/PD-L1 expression both on immune cells and on tumor cells, and potential correlations between the PD-1/PD-L1 axis and tumor hypoxia dynamics during chemoradiation were assessed using Spearman's rank correlations. Hypoxia dynamics during treatment were quantified by subtracting the standardized uptake value (SUV) index at baseline from the SUV values at weeks 2 or 5, whereby SUV index was defined as ratio of maximum tumor [18F]FMISO SUV to mean SUV in the contralateral sternocleidomastoid muscle (i.e. tumor-to-muscle ratio). The impact of the PD-1/PD-L1 expression alone and in combination with persistent tumor hypoxia on locoregional control (LRC), progression-free survival (PFS) and overall survival (OS) was examined using log-rank tests and Cox proportional hazards models. Results: Neither PD-L1 nor PD-1 expression levels on tumor-infiltrating immune cells influenced LRC (HR = 0.734; p = 0.480 for PD-L1, HR = 0.991; p = 0.989 for PD-1), PFS (HR = 0.813; p = 0.597 for PD-L1, HR = 0.796; p = 0.713 for PD-1) or OS (HR = 0.698; p = 0.405 for PD-L1, HR = 0.315; p = 0.265 for PD-1). However, patients with no hypoxia resolution between weeks 0 and 2 and PD-L1 expression on tumor cells, quantified by a tumor proportional score (TPS) of at least 1%, showed significantly worse LRC (HR = 3.374, p = 0.022) and a trend towards reduced PFS (HR = 2.752, p = 0.052). In the multivariate Cox regression analysis, the combination of absent tumor hypoxia resolution and high tumoral PD-L1 expression remained a significant prognosticator for impaired LRC (HR = 3.374, p = 0.022). On the other side, tumoral PD-L1 expression did not compromise the outcomes of patients whose tumor-associated hypoxia declined between week 0 and 2 during chemoradiation (LRC: HR = 1.186, p = 0.772, PFS: HR = 0.846, p = 0.766). Conclusion: In this exploratory analysis, we showed for the first time that patients with both persistent tumor-associated hypoxia during treatment and PD-L1 expression on tumor cells exhibited a worse outcome, while the tumor cells' PD-L1 expression did not influence the outcomes of patients with early tumor hypoxia resolution. While the results have to be validated in an independent cohort, these findings form a foundation to investigate the combination of hypoxic modification and immune checkpoint inhibitors for the unfavorable subgroup, moving forward towards personalized radiation oncology treatment.
Collapse
|