1
|
Meunier M, Yammine A, Bettaieb A, Plenchette S. Nitroglycerin: a comprehensive review in cancer therapy. Cell Death Dis 2023; 14:323. [PMID: 37173331 PMCID: PMC10182021 DOI: 10.1038/s41419-023-05838-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Nitroglycerin (NTG) is a prodrug that has long been used in clinical practice for the treatment of angina pectoris. The biotransformation of NTG and subsequent release of nitric oxide (NO) is responsible for its vasodilatating property. Because of the remarkable ambivalence of NO in cancer disease, either protumorigenic or antitumorigenic (partly dependent on low or high concentrations), harnessing the therapeutic potential of NTG has gain interest to improve standard therapies in oncology. Cancer therapeutic resistance remains the greatest challenge to overcome in order to improve the management of cancer patients. As a NO releasing agent, NTG has been the subject of several preclinical and clinical studies used in combinatorial anticancer therapy. Here, we provide an overview of the use of NTG in cancer therapy in order to foresee new potential therapeutic avenues.
Collapse
Affiliation(s)
- Mélina Meunier
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université de Bourgogne, Dijon, France
- LIIC, EPHE, PSL Research University, Paris, France
| | - Aline Yammine
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université de Bourgogne, Dijon, France
- LIIC, EPHE, PSL Research University, Paris, France
| | - Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université de Bourgogne, Dijon, France
- LIIC, EPHE, PSL Research University, Paris, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers (LIIC), EA7269, Université de Bourgogne, Dijon, France.
- LIIC, EPHE, PSL Research University, Paris, France.
| |
Collapse
|
2
|
He L, Li Z, Chen X, Huang Y, Yan L, Liang C, Liu Z. A radiomics prognostic scoring system for predicting progression-free survival in patients with stage IV non-small cell lung cancer treated with platinum-based chemotherapy. Chin J Cancer Res 2021; 33:592-605. [PMID: 34815633 PMCID: PMC8580802 DOI: 10.21147/j.issn.1000-9604.2021.05.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/08/2021] [Indexed: 12/23/2022] Open
Abstract
Objective To develop and validate a radiomics prognostic scoring system (RPSS) for prediction of progression-free survival (PFS) in patients with stage IV non-small cell lung cancer (NSCLC) treated with platinum-based chemotherapy. Methods In this retrospective study, four independent cohorts of stage IV NSCLC patients treated with platinum-based chemotherapy were included for model construction and validation (Discovery: n=159; Internal validation: n=156; External validation: n=81, Mutation validation: n=64). First, a total of 1,182 three-dimensional radiomics features were extracted from pre-treatment computed tomography (CT) images of each patient. Then, a radiomics signature was constructed using the least absolute shrinkage and selection operator method (LASSO) penalized Cox regression analysis. Finally, an individualized prognostic scoring system incorporating radiomics signature and clinicopathologic risk factors was proposed for PFS prediction. Results The established radiomics signature consisting of 16 features showed good discrimination for classifying patients with high-risk and low-risk progression to chemotherapy in all cohorts (All P<0.05). On the multivariable analysis, independent factors for PFS were radiomics signature, performance status (PS), and N stage, which were all selected into construction of RPSS. The RPSS showed significant prognostic performance for predicting PFS in discovery [C-index: 0.772, 95% confidence interval (95% CI): 0.765−0.779], internal validation (C-index: 0.738, 95% CI: 0.730−0.746), external validation (C-index: 0.750, 95% CI: 0.734−0.765), and mutation validation (C-index: 0.739, 95% CI: 0.720−0.758). Decision curve analysis revealed that RPSS significantly outperformed the clinicopathologic-based model in terms of clinical usefulness (All P<0.05). Conclusions This study established a radiomics prognostic scoring system as RPSS that can be conveniently used to achieve individualized prediction of PFS probability for stage IV NSCLC patients treated with platinum-based chemotherapy, which holds promise for guiding personalized pre-therapy of stage IV NSCLC.
Collapse
Affiliation(s)
- Lan He
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zhenhui Li
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming 650118, China
| | - Xin Chen
- Department of Radiology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510120, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Lixu Yan
- Department of Pathology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| |
Collapse
|
3
|
Schaner PE, Williams BB, Chen EY, Pettus JR, Schreiber WA, Kmiec MM, Jarvis LA, Pastel DA, Zuurbier RA, DiFlorio-Alexander RM, Paydarfar JA, Gosselin BJ, Barth RJ, Rosenkranz KM, Petryakov SV, Hou H, Tse D, Pletnev A, Flood AB, Wood VA, Hebert KA, Mosher RE, Demidenko E, Swartz HM, Kuppusamy P. First-In-Human Study in Cancer Patients Establishing the Feasibility of Oxygen Measurements in Tumors Using Electron Paramagnetic Resonance With the OxyChip. Front Oncol 2021; 11:743256. [PMID: 34660306 PMCID: PMC8517507 DOI: 10.3389/fonc.2021.743256] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/07/2021] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE The overall objective of this clinical study was to validate an implantable oxygen sensor, called the 'OxyChip', as a clinically feasible technology that would allow individualized tumor-oxygen assessments in cancer patients prior to and during hypoxia-modification interventions such as hyperoxygen breathing. METHODS Patients with any solid tumor at ≤3-cm depth from the skin-surface scheduled to undergo surgical resection (with or without neoadjuvant therapy) were considered eligible for the study. The OxyChip was implanted in the tumor and subsequently removed during standard-of-care surgery. Partial pressure of oxygen (pO2) at the implant location was assessed using electron paramagnetic resonance (EPR) oximetry. RESULTS Twenty-three cancer patients underwent OxyChip implantation in their tumors. Six patients received neoadjuvant therapy while the OxyChip was implanted. Median implant duration was 30 days (range 4-128 days). Forty-five successful oxygen measurements were made in 15 patients. Baseline pO2 values were variable with overall median 15.7 mmHg (range 0.6-73.1 mmHg); 33% of the values were below 10 mmHg. After hyperoxygenation, the overall median pO2 was 31.8 mmHg (range 1.5-144.6 mmHg). In 83% of the measurements, there was a statistically significant (p ≤ 0.05) response to hyperoxygenation. CONCLUSIONS Measurement of baseline pO2 and response to hyperoxygenation using EPR oximetry with the OxyChip is clinically feasible in a variety of tumor types. Tumor oxygen at baseline differed significantly among patients. Although most tumors responded to a hyperoxygenation intervention, some were non-responders. These data demonstrated the need for individualized assessment of tumor oxygenation in the context of planned hyperoxygenation interventions to optimize clinical outcomes.
Collapse
Affiliation(s)
- Philip E. Schaner
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Benjamin B. Williams
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Eunice Y. Chen
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Jason R. Pettus
- Department of Pathology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Wilson A. Schreiber
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Maciej M. Kmiec
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Lesley A. Jarvis
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - David A. Pastel
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Rebecca A. Zuurbier
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Roberta M. DiFlorio-Alexander
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Joseph A. Paydarfar
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Benoit J. Gosselin
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Richard J. Barth
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Kari M. Rosenkranz
- Department of Surgery, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Sergey V. Petryakov
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Huagang Hou
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Dan Tse
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Alexandre Pletnev
- Department of Chemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Ann Barry Flood
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Victoria A. Wood
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Kendra A. Hebert
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Robyn E. Mosher
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Eugene Demidenko
- Department of Biomedical Data Science, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Harold M. Swartz
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Periannan Kuppusamy
- Department of Medicine, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Radiology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
- Department of Chemistry, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth College, and Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
4
|
Tomaszewski MR, Gillies RJ. The Biological Meaning of Radiomic Features. Radiology 2021; 298:505-516. [PMID: 33399513 PMCID: PMC7924519 DOI: 10.1148/radiol.2021202553] [Citation(s) in RCA: 249] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/30/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023]
Abstract
An earlier incorrect version appeared online. This article was corrected on February 10, 2021.
Collapse
Affiliation(s)
- Michal R. Tomaszewski
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| | - Robert J. Gillies
- From the Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Dr, Tampa, FL 33612
| |
Collapse
|
5
|
Park S, Lee Y, Kim TS, Kim SK, Han JY. Response evaluation after immunotherapy in NSCLC: Early response assessment using FDG PET/CT. Medicine (Baltimore) 2020; 99:e23815. [PMID: 33371161 PMCID: PMC7748304 DOI: 10.1097/md.0000000000023815] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 11/19/2020] [Indexed: 11/26/2022] Open
Abstract
The present study aimed to evaluate the role of early F-18 2-deoxy-2-[fluorine-18] fluoro-D-glucose positron emission tomography/computed tomography (FDG PET/CT) in non-small cell lung cancer patients undergoing immune checkpoint inhibitor (ICI) treatment.Twenty-four non-small cell lung cancer patients who received nivolumab or pembrolizumab and underwent FDG PET/CT as an interim analysis after 2 or 3 cycles of ICI treatment were retrospectively enrolled. Tumor response was assessed using the PET Response Criteria in Solid Tumors 1.0 (PERCIST) and the European Organization for Research and Treatment of Cancer (EORTC) criteria after 2 or 3 cycles of ICI treatment (SCAN-1) and after an additional 2 cycles of ICI treatment (SCAN-2). The best overall response was determined by FDG PET/CT or chest CT at ≥ 3 months after therapy initiation, and the clinical benefit was investigated. progression-free survival was investigated, and its correlation with clinicopathologic and metabolic parameters was examined using a Cox multivariate proportional hazards model.In the interim analysis, 4 patients achieved a complete metabolic response (CMR), 1 patient exhibited a partial metabolic response (PMR), and 14 patients had Progressive metabolic disease (PMD) according to the PERCIST and EORTC criteria. Four patients showed stable metabolic disease (SMD) according to the PERCIST criteria, and 2 patients showed different responses (i.e., PMR) according to the EORTC criteria. Patients with a CMR or PMR at SCAN-1 had a clinical benefit. Among the 4 patients with SMD at SCAN-1, only 1 experienced a clinical benefit regardless of the percent change in the peak standardized uptake value. Two patients with discordant response assessments between the PERCIST and EORTC criteria showed conflicting clinical benefits. Among the 14 patients with PMD, none experienced any clinical benefit. Only metabolic parameters were significant factors for predicting progression in the multivariate analysis (peak standardized uptake value and metabolic tumor volume, HRs of 1.18 and 1.00, respectively).Based on early F-18 FDG PET/CT after ICI treatment, metabolic parameters could predict post-treatment progression. Responses after ICI treatment were correctly assessed in patients with a CMR, a PMR, and PMD, but patients with SMD required a meticulous follow-up because of varying clinical benefits.
Collapse
Affiliation(s)
- Sohyun Park
- Department of Nuclear Medicine, Guro Hospital, Korea University College of Medicine, Goyang, Republic of Korea
- Department of Nuclear Medicine
| | | | | | - Seok-ki Kim
- Department of Nuclear Medicine
- Molecular Imaging Branch, Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Korea
| | | |
Collapse
|
6
|
Vong LB, Nagasaki Y. Nitric Oxide Nano-Delivery Systems for Cancer Therapeutics: Advances and Challenges. Antioxidants (Basel) 2020; 9:E791. [PMID: 32858970 PMCID: PMC7555477 DOI: 10.3390/antiox9090791] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/21/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) plays important roles in various physiological and pathological functions and processes in the human body. Therapeutic application of NO molecules has been investigated in various diseases, including cardiovascular disease, cancer, and infections. However, the extremely short half-life of NO, which limits its clinical use considerably, along with non-specific distribution, has resulted in a low therapeutic index and undesired adverse effects. To overcome the drawbacks of using this gaseous signaling molecule, researchers in the last several decades have focused on innovative medical technologies, specifically nanoparticle-based drug delivery systems (DDSs), because these systems alter the biodistribution of the therapeutic agent through controlled release at the target tissues, resulting in a significant therapeutic drug effect. Thus, the application of nano-systems for NO delivery in the field of biomedicine, particularly in the development of new drugs for cancer treatment, has been increasing worldwide. In this review, we discuss NO delivery nanoparticle systems, with the aim of improving drug delivery development for conventional chemotherapies and controlling multidrug resistance in cancer treatments.
Collapse
Affiliation(s)
- Long Binh Vong
- School of Biomedical Engineering, International University, Ho Chi Minh 700000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh 700000, Vietnam
| | - Yukio Nagasaki
- Department of Materials Science, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8573, Japan
- Master’s School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8577, Japan
- Center for Research in Isotopes and Environmental Dynamics (CRiED), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan
| |
Collapse
|
7
|
Liu Y. Application of artificial intelligence in clinical non-small cell lung cancer. Artif Intell Cancer 2020; 1:19-30. [DOI: 10.35713/aic.v1.i1.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 02/06/2023] Open
Abstract
Lung cancer is the most common cause of cancer death in the world. Early diagnosis, screening and precise individualized treatment can significantly reduce the death rate of lung cancer. Artificial intelligence (AI) has been shown to be able to help clinicians make more accurate judgments and decisions in many ways. It has been involved in the screening of lung cancer, the judgment of benign and malignant degree of pulmonary nodules, the classification of histological cancer, the differentiation of histological subtypes, the identification of genomics, the judgment of the effectiveness of treatment and even the prognosis. AI has shown that it can be an excellent assistant for clinicians. This paper reviews the application of AI in the field of non-small cell lung cancer and describes the relevant progress. Although most of the studies to evaluate the clinical application of AI in non-small cell lung cancer have not been repeatable and generalizable, the research results highlight the efforts to promote the clinical application of AI technology and influence the future treatment direction.
Collapse
Affiliation(s)
- Yong Liu
- Department of Thoracic Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430011, Hubei Province, China
| |
Collapse
|
8
|
|
9
|
Can radiomics help to predict skeletal muscle response to chemotherapy in stage IV non-small cell lung cancer? Eur J Cancer 2019; 120:107-113. [PMID: 31514107 DOI: 10.1016/j.ejca.2019.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/24/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Muscle depletion negatively impacts treatment efficacy and survival rates in cancer. Prevention and timely treatment of muscle loss require prediction of patients at risk. We aimed to investigate the potential of skeletal muscle radiomic features to predict future muscle loss. METHODS A total of 116 patients with stage IV non-small cell lung cancer included in a randomised controlled trial (NCT01171170) studying the effect of nitroglycerin added to paclitaxel-carboplatin-bevacizumab were enrolled. In this post hoc analysis, muscle cross-sectional area and radiomic features were extracted from computed tomography images obtained before initiation of chemotherapy and shortly after administration of the second cycle. For internal cross-validation, the cohort was randomly split in a training set and validation set 100 times. We used least absolute shrinkage and selection operator method to select features that were most significantly associated with muscle loss and an area under the curve (AUC) for model performance. RESULTS Sixty-nine patients (59%) exhibited loss of skeletal muscle. One hundred ninety-three features were used to construct a prediction model for muscle loss. The average AUC was 0.49 (95% confidence interval [CI]: 0.36, 0.62). Differences in intensity and texture radiomic features over time were seen between patients with and without muscle loss. CONCLUSIONS The present study shows that skeletal muscle radiomics did not predict future muscle loss during chemotherapy in non-small cell lung cancer. Differences in radiomic features over time might reflect myosteatosis. Future imaging analysis combined with muscle tissue analysis in patients and in experimental models is needed to unravel the biological processes linked to the radiomic features.
Collapse
|
10
|
Degens JHRJ, Sanders KJC, de Jong EEC, Groen HJM, Smit EF, Aerts JG, Schols AMWJ, Dingemans AMC. The prognostic value of early onset, CT derived loss of muscle and adipose tissue during chemotherapy in metastatic non-small cell lung cancer. Lung Cancer 2019; 133:130-135. [PMID: 31200819 DOI: 10.1016/j.lungcan.2019.05.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/13/2019] [Accepted: 05/17/2019] [Indexed: 12/25/2022]
Abstract
OBJECTIVES To evaluate the relationship between early changes in muscle and adipose tissue during chemotherapy and overall survival (OS) in stage IV non-small cell lung cancer (NSCLC). MATERIALS AND METHODS In this post-hoc analysis of the first line NVALT12 trial (NCT01171170) in stage IV NSCLC, skeletal muscle (SM), radiation attenuation (RA), subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) were assessed at the third lumbar level on CT-images obtained before initiation of chemotherapy and shortly after administration of the second cycle. The contribution of changes in different body compartments to overall survival was assessed. RESULTS CT scans of 111 patients were included. Analysis of body composition changes between the baseline and the follow-up scan, revealed that overall SM cross sectional area (CSA), radiation attenuation and SAT CSA decreased respectively by -1.2 ± 2.9 cm2/m2 (p < 0.001), -0.7 ± 3.3 HU (p = 0.026) and -1.9 ± 8.7 cm2/m2 (p = 0.026), while no significant changes in VAT tissue were observed. Longitudinally, median OS was significantly shorter among patients losing SM compared to patients with preserved SM (9.4 versus 14.2 months; HR 1.9, 95% CI: 1.23, 2.79, p = 0.003). Multivariate analyses showed that proportional loss of muscle mass was associated with poor OS (HR 0.949, 95% CI: 0.915, 0.985, p = 0.006) independent from important clinical prognostic factors including WHO-PS, gender, age and Charlson comorbidity index. CONCLUSION Early loss of SM during first line chemotherapy is a poor prognostic factor in stage IV NSCLC patients. Future studies have to reveal whether early supportive intervention guided by initial CT muscle response to chemotherapy can influence the wasting process and related mortality risk.
Collapse
Affiliation(s)
- J H R J Degens
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - K J C Sanders
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - E E C de Jong
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - H J M Groen
- Department of Respiratory Medicine, University Medical Center Groningen, Groningen, the Netherlands
| | - E F Smit
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - J G Aerts
- Department of Respiratory Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - A M W J Schols
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - A-M C Dingemans
- Department of Respiratory Medicine, GROW School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|
11
|
Zhang L, Liang Y, Li S, Zeng F, Meng Y, Chen Z, Liu S, Tao Y, Yu F. The interplay of circulating tumor DNA and chromatin modification, therapeutic resistance, and metastasis. Mol Cancer 2019; 18:36. [PMID: 30849971 PMCID: PMC6408771 DOI: 10.1186/s12943-019-0989-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Peripheral circulating free DNA (cfDNA) is DNA that is detected in plasma or serum fluid with a cell-free status. For cancer patients, cfDNA not only originates from apoptotic cells but also from necrotic tumor cells and disseminated tumor cells that have escaped into the blood during epithelial-mesenchymal transition. Additionally, cfDNA derived from tumors, also known as circulating tumor DNA (ctDNA), carries tumor-associated genetic and epigenetic changes in cancer patients, which makes ctDNA a potential biomarker for the early diagnosis of tumors, monitory and therapeutic evaluations, and prognostic assessments, among others, for various kinds of cancer. Moreover, analyses of cfDNA chromatin modifications can reflect the heterogeneity of tumors and have potential for predicting tumor drug resistance.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yiyi Liang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shifu Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Fanyuan Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongan Meng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ziwei Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China.
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
12
|
Bi WL, Hosny A, Schabath MB, Giger ML, Birkbak NJ, Mehrtash A, Allison T, Arnaout O, Abbosh C, Dunn IF, Mak RH, Tamimi RM, Tempany CM, Swanton C, Hoffmann U, Schwartz LH, Gillies RJ, Huang RY, Aerts HJWL. Artificial intelligence in cancer imaging: Clinical challenges and applications. CA Cancer J Clin 2019; 69:127-157. [PMID: 30720861 PMCID: PMC6403009 DOI: 10.3322/caac.21552] [Citation(s) in RCA: 661] [Impact Index Per Article: 132.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Judgement, as one of the core tenets of medicine, relies upon the integration of multilayered data with nuanced decision making. Cancer offers a unique context for medical decisions given not only its variegated forms with evolution of disease but also the need to take into account the individual condition of patients, their ability to receive treatment, and their responses to treatment. Challenges remain in the accurate detection, characterization, and monitoring of cancers despite improved technologies. Radiographic assessment of disease most commonly relies upon visual evaluations, the interpretations of which may be augmented by advanced computational analyses. In particular, artificial intelligence (AI) promises to make great strides in the qualitative interpretation of cancer imaging by expert clinicians, including volumetric delineation of tumors over time, extrapolation of the tumor genotype and biological course from its radiographic phenotype, prediction of clinical outcome, and assessment of the impact of disease and treatment on adjacent organs. AI may automate processes in the initial interpretation of images and shift the clinical workflow of radiographic detection, management decisions on whether or not to administer an intervention, and subsequent observation to a yet to be envisioned paradigm. Here, the authors review the current state of AI as applied to medical imaging of cancer and describe advances in 4 tumor types (lung, brain, breast, and prostate) to illustrate how common clinical problems are being addressed. Although most studies evaluating AI applications in oncology to date have not been vigorously validated for reproducibility and generalizability, the results do highlight increasingly concerted efforts in pushing AI technology to clinical use and to impact future directions in cancer care.
Collapse
Affiliation(s)
- Wenya Linda Bi
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Ahmed Hosny
- Research Scientist, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Matthew B. Schabath
- Associate Member, Department of Cancer EpidemiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Maryellen L. Giger
- Professor of Radiology, Department of RadiologyUniversity of ChicagoChicagoIL
| | - Nicolai J. Birkbak
- Research Associate, The Francis Crick InstituteLondonUnited Kingdom
- Research Associate, University College London Cancer InstituteLondonUnited Kingdom
| | - Alireza Mehrtash
- Research Assistant, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Research Assistant, Department of Electrical and Computer EngineeringUniversity of British ColumbiaVancouverBCCanada
| | - Tavis Allison
- Research Assistant, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Research Assistant, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Omar Arnaout
- Assistant Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Christopher Abbosh
- Research Fellow, The Francis Crick InstituteLondonUnited Kingdom
- Research Fellow, University College London Cancer InstituteLondonUnited Kingdom
| | - Ian F. Dunn
- Associate Professor of Neurosurgery, Department of Neurosurgery, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Raymond H. Mak
- Associate Professor, Department of Radiation Oncology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Rulla M. Tamimi
- Associate Professor, Department of MedicineBrigham and Women’s Hospital, Dana‐Farber Cancer Institute, Harvard Medical SchoolBostonMA
| | - Clare M. Tempany
- Professor of Radiology, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Charles Swanton
- Professor, The Francis Crick InstituteLondonUnited Kingdom
- Professor, University College London Cancer InstituteLondonUnited Kingdom
| | - Udo Hoffmann
- Professor of Radiology, Department of RadiologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA
| | - Lawrence H. Schwartz
- Professor of Radiology, Department of RadiologyColumbia University College of Physicians and SurgeonsNew YorkNY
- Chair, Department of RadiologyNew York Presbyterian HospitalNew YorkNY
| | - Robert J. Gillies
- Professor of Radiology, Department of Cancer PhysiologyH. Lee Moffitt Cancer Center and Research InstituteTampaFL
| | - Raymond Y. Huang
- Assistant Professor, Department of Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
| | - Hugo J. W. L. Aerts
- Associate Professor, Departments of Radiation Oncology and Radiology, Brigham and Women’s Hospital, Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMA
- Professor in AI in Medicine, Radiology and Nuclear Medicine, GROWMaastricht University Medical Centre (MUMC+)MaastrichtThe Netherlands
| |
Collapse
|
13
|
Xie X, Chen H, Yang H, Lin H, Zhou S, Shen R, Lu C, Ling L, Lin W, Liao Z. Predictive value of positron emission tomography for the prognosis of molecularly targeted therapy in solid tumors. Onco Targets Ther 2018; 11:8885-8899. [PMID: 30573975 PMCID: PMC6290871 DOI: 10.2147/ott.s178076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objective This study aimed at comprehensively exploring the value applying positron emission tomography (PET) to predict the effect of molecularly targeted therapy in solid tumors. Materials and methods A systematic search was performed for potentially relevant studies from the time of inception to February 2017. The primary endpoints were progression-free survival (PFS), overall survival (OS), and time to progression (TTP). The results were analyzed by Review Manager version 5.3 (RevMan 5.3) statistical software. Subgroup analyses were implemented based on the type of molecularly targeted agents (monoclonal antibodies arm and small molecular targeted agents arm), mechanism (erlotinib/gefitinib arm and bevacizumab arm), radioactive tracers, type of tumor, and reevaluated PET timing. Results Twenty-six studies incorporating 865 individuals were eligible. Compared with PET nonresponse group, PET response group displayed a decrease in maximal standard uptake value (SUVmax), which was associated with a significantly prolonged PFS (HR =0.41, 95% CI [0.29, 0.59]; P<0.00001), OS (HR =0.52, 95% CI [0.40, 0.67]; P<0.00001), and TTP (HR =0.30, 95% CI [0.14, 0.66]; P=0.003). Similar results were obtained in the subgroup analyses of PFS in erlotinib/gefitinib arm and small molecular targeted agents arm; and OS in lung cancer arm, erlotinib/gefitinib arm, bevacizumab arm, small molecular targeted agents arm, monoclonal antibodies arm, 18F-fluorodeoxythymidine (18F-FLT) arm, 18F-fluorodeoxyglucose (18F-FDG) arm, and early PET timing arm. Conclusion Our study demonstrated that PET was a favorable approach to predict the prognosis of molecularly targeted therapy for solid tumors. PET assessment within 2 weeks could be useful to predict clinical outcome.
Collapse
Affiliation(s)
- Xianhe Xie
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Huijuan Chen
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Haitao Yang
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Heng Lin
- Department of Oncology, Fuzhou Pulmonary Hospital, Fuzhou, Fujian, People's Republic of China
| | - Sijing Zhou
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Ruifen Shen
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Cuiping Lu
- Department of Medical Oncology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, Fujian, People's Republic of China
| | - Liting Ling
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Wanzun Lin
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| | - Ziyuan Liao
- Department of Chemotherapy, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China,
| |
Collapse
|
14
|
The role of functional imaging in lung cancer. Clin Transl Imaging 2018. [DOI: 10.1007/s40336-018-0300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
15
|
Integrated texture parameter of 18F-FDG PET may be a stratification factor for the survival of nonoperative patients with locally advanced non-small-cell lung cancer. Nucl Med Commun 2018; 39:732-740. [DOI: 10.1097/mnm.0000000000000860] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Shaikh F, Franc B, Allen E, Sala E, Awan O, Hendrata K, Halabi S, Mohiuddin S, Malik S, Hadley D, Shrestha R. Translational Radiomics: Defining the Strategy Pipeline and Considerations for Application-Part 2: From Clinical Implementation to Enterprise. J Am Coll Radiol 2018; 15:543-549. [PMID: 29366598 DOI: 10.1016/j.jacr.2017.12.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 12/07/2017] [Indexed: 12/18/2022]
Abstract
Enterprise imaging has channeled various technological innovations to the field of clinical radiology, ranging from advanced imaging equipment and postacquisition iterative reconstruction tools to image analysis and computer-aided detection tools. More recently, the advancement in the field of quantitative image analysis coupled with machine learning-based data analytics, classification, and integration has ushered in the era of radiomics, a paradigm shift that holds tremendous potential in clinical decision support as well as drug discovery. However, there are important issues to consider to incorporate radiomics into a clinically applicable system and a commercially viable solution. In this two-part series, we offer insights into the development of the translational pipeline for radiomics from methodology to clinical implementation (Part 1) and from that point to enterprise development (Part 2). In Part 2 of this two-part series, we study the components of the strategy pipeline, from clinical implementation to building enterprise solutions.
Collapse
Affiliation(s)
- Faiq Shaikh
- Institute of Computational Health Sciences, UCSF, San Francisco, California.
| | - Benjamin Franc
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California
| | | | - Evis Sala
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Omer Awan
- Department of Radiology, Temple University, Philadelphia, Pennsylvania
| | | | - Safwan Halabi
- Department of Radiology, Stanford University, Palo Alto, California
| | - Sohaib Mohiuddin
- Department of Radiology, Division of Nuclear Medicine, University of Miami, Miami, Florida
| | - Sana Malik
- School of Social Welfare, Stony Brook University, New York, New York
| | - Dexter Hadley
- Institute of Computational Health Sciences, UCSF, San Francisco, California
| | | |
Collapse
|
17
|
de Jong EEC, van Elmpt W, Hoekstra OS, Groen HJM, Smit EF, Boellaard R, Lambin P, Dingemans AMC. Quality assessment of positron emission tomography scans: recommendations for future multicentre trials. Acta Oncol 2017; 56:1459-1464. [PMID: 28830270 DOI: 10.1080/0284186x.2017.1346824] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Standardization protocols and guidelines for positron emission tomography (PET) in multicenter trials are available, despite a large variability in image acquisition and reconstruction parameters exist. In this study, we investigated the compliance of PET scans to the guidelines of the European Association of Nuclear Medicine (EANM). From these results, we provide recommendations for future multicenter studies using PET. MATERIAL AND METHODS Patients included in a multicenter randomized phase II study had repeated PET scans for early response assessment. Relevant acquisition and reconstruction parameters were extracted from the digital imaging and communications in medicine (DICOM) header of the images. The PET image parameters were compared to the guidelines of the EANM for tumor imaging version 1.0 recommended parameters. RESULTS From the 223 included patients, 167 baseline scans and 118 response scans were available from 15 hospitals. Scans of 19% of the patients had an uptake time that fulfilled the Uniform Protocols for Imaging in Clinical Trials response assessment criteria. The average quality score over all hospitals was 69%. Scans with a non-compliant uptake time had a larger standard deviation of the mean standardized uptake value (SUVmean) of the liver than scans with compliant uptake times. CONCLUSIONS Although a standardization protocol was agreed on, there was a large variability in imaging parameters. For future, multicenter studies including PET imaging a prospective central quality review during patient inclusion is needed to improve compliance with image standardization protocols as defined by EANM.
Collapse
Affiliation(s)
- Evelyn E. C. de Jong
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Wouter van Elmpt
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Otto S. Hoekstra
- Department of Nuclear Medicine & PET Research, VU University Medical Center, Amsterdam, Netherlands
| | - Harry J. M. Groen
- Department of Pulmonary Diseases, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Egbert F. Smit
- Department of Pulmonary Diseases, VU University Medical Center, Amsterdam, Netherlands
- Department of Thoracic Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Philippe Lambin
- Department of Radiation Oncology (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| | - Anne-Marie C. Dingemans
- Department of Pulmonology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, Netherlands
| |
Collapse
|