1
|
Ceriani L, Milan L, Chauvie S, Zucca E. Understandings 18 FDG PET radiomics and its application to lymphoma. Br J Haematol 2025. [PMID: 40230306 DOI: 10.1111/bjh.20074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025]
Abstract
The early identification of lymphoma patients who fail front-line treatment is crucial for optimizing disease management. Positron emission tomography, a well-established tool for staging and response evaluation in lymphoma, is typically assessed visually or semiquantitatively, leaving much of its latent information unexploited. Radiomic analysis, which employs mathematical descriptors, can enable the extraction of quantitative features from baseline images that correlate with the disease's biological characteristics. Emerging radiomic features such as metabolic tumour volume, total lesion glycolysis and markers of disease dissemination and metabolic heterogeneity are proving to be powerful prognostic biomarkers in lymphoma. Texture analysis, the most advanced area of radiomics, offers highly complex features that require further standardization and validation before being adopted as reliable biomarkers. Combining radiomic features with clinical risk factors and genomic data holds promising potential for improving clinical risk prediction. This review explores the current state of radiomic analysis, progress towards its standardization and its incorporation into clinical practice and trial designs. The integration of radiomic markers with circulating tumour DNA may provide a comprehensive approach to developing baseline and dynamic risk scores, facilitating the testing of novel treatments and advancing personalized treatment of aggressive lymphomas.
Collapse
Affiliation(s)
- Luca Ceriani
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Lisa Milan
- Nuclear Medicine and PET/CT Centre, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Stephane Chauvie
- Medical Physics Division, Santa Croce e Carlo Hospital, Cuneo, Italy
| | - Emanuele Zucca
- Institute of Oncology Research, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Haematology, Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Medical Oncology, Bern University Hospital and University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Shi L, Li C, Bai Y, Cao Y, Zhao S, Chen X, Cheng Z, Zhang Y, Li H. CT radiomics to predict pathologic complete response after neoadjuvant immunotherapy plus chemoradiotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2025; 35:1594-1604. [PMID: 39470794 DOI: 10.1007/s00330-024-11141-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE To develop and validate a CT-based radiomics model to predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC). METHODS A total of 105 patients with locally advanced ESCC receiving NICRT from February 2019 to December 2023 were enrolled. Patients were randomly divided into the training cohort and the test cohort at a 3:1 ratio. Enhanced CT scans were obtained before NICRT treatment. The 2D and 3D regions of interest were segmented, and features were extracted, followed by feature selection. Six algorithms were applied to construct the radiomics and clinical models. These models were evaluated by area under curve (AUC), accuracy, sensitivity, and specificity, and their respective optimal algorithms were further compared. RESULTS Forty-eight patients (45.75%) achieved pCR after NICRT. The AUC values of three algorithms in 2D radiomics models were higher than those in the 3D radiomics model and clinical model. Among these, the 2D radiomics model based on eXtreme Gradient Boosting (XGBoost) exhibited the best performance, with an AUC of 0.89 (95% CI, 0.81-0.97), accuracy of 0.85, sensitivity of 0.86, and specificity of 0.84 in the training cohort, and an AUC of 0.80 (95% CI, 0.64-0.97), accuracy of 0.77, sensitivity of 0.84, and specificity of 0.69 in the test cohort. Calibration curves also showed good agreement between predicted and actual response, and the decision curve analysis further confirmed its clinical applicability. CONCLUSION The 2D radiomics model can effectively predict pCR to NICRT in locally advanced ESCC. KEY POINTS Question Can CT-based radiomics predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC)? Findings The model based on eXtreme Gradient Boosting (XGBoost) performed best, with an AUC of 0.89 in the training and 0.80 in the test cohort. Clinical relevance This CT-based radiomics model exhibits promising performance for predicting pCR to NICRT in locally advanced ESCC, which may be valuable in personalized treatment plan optimization.
Collapse
Affiliation(s)
- Liqiang Shi
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yaya Bai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqin Cao
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengguang Zhao
- Department of Radiotherapy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zenghui Cheng
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajie Zhang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
3
|
Alberts IL, Xue S, Sari H, Cavinato L, Prenosil G, Afshar-Oromieh A, Mingels C, Shi K, Caobelli F, Rahmim A, Pyka T, Rominger A. Long-axial field-of-view PET/CT improves radiomics feature reliability. Eur J Nucl Med Mol Imaging 2025; 52:1004-1016. [PMID: 39477863 DOI: 10.1007/s00259-024-06921-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 09/11/2024] [Indexed: 01/23/2025]
Abstract
PURPOSE To assess the influence of long-axial field-of-view (LAFOV) PET/CT systems on radiomics feature reliability, to assess the suitability for short-duration or low-activity acquisitions for textural feature analysis and to investigate the influence of acceptance angle. METHODS 34 patients were analysed: twelve patients underwent oncological 2-[18F]-FDG PET/CT, fourteen [18F]PSMA-1007 and eight [68Ga]Ga-DOTATOC. Data were obtained using a 106 cm LAFOV system for 10 min. Sinograms were generated from list-mode data corresponding to scan durations of 2, 5, 10, 20, 30, 60, 120, 240, 360 and 600s using both standard (minimum ring difference MRD 85 crystals) and maximum acceptance angles (MRD 322). Target lesions were segmented and radiomics features were calculated. To assess feature correlation, Pearson's product-moment correlation coefficient (PPMCC) was calculated with respect to the full duration acquisition for MRD 85 and 322 respectively. The number of features with excellent (r > 0.9), moderate (r > 0.7 and < 0.9) and poor (r ≤ 0.7) correlation was compared as a measure of feature stability. Intra-class heterogeneity was assessed by means of the quartile coefficient of dispersion. RESULTS As expected, PPMCC improved with acquisition time for all features. By 240s almost all features showed at least moderate agreement with the full count (C100%) data, and by 360s almost all showed excellent agreement. Compared to standard-axial field of view (SAFOV) equivalent scans, fewer features showed moderate or poor agreement, and this was most pronounced for [68Ga]Ga-DOTATOC. Data obtained at C100% at MRD 322 were better able to capture between-patient heterogeneities. CONCLUSION The improved feature reliability at longer acquisition times and higher MRD demonstrate the advantages of high sensitivity LAFOV systems for reproducible and low-noise data. High fidelity between MRD 85 and MRD 322 was seen at all scan durations > 2s. When contrasted with data comparable to a simulated SAFOV acquisition, full-count and full-MRD data were better able to capture underlying feature heterogeneities.
Collapse
Affiliation(s)
- Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer - Vancouver, 600 West 10th Ave, Vancouver, BC, V5Z 1H5, Canada.
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland.
| | - Song Xue
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland
| | - Lara Cavinato
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
- Laboratory for Modelling and Scientific Computing, Department of Mathematics, Politecnico di Milano, Milan, 20133, Italy
| | - George Prenosil
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Federico Caobelli
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Arman Rahmim
- Molecular Imaging and Therapy, BC Cancer - Vancouver, 600 West 10th Ave, Vancouver, BC, V5Z 1H5, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, V5Z 1M9, Canada
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern, Bern, Switzerland
| |
Collapse
|
4
|
Duffles G, Maués JHDS, Lupinacci F, Pereira LG, Ferreira EN, Freitas L, Niemann F, Takahashi MES, Ramos CD, Chauffaille MDLLF, Lorand-Metze I. Circulating tumor DNA in diffuse large B-cell lymphoma: analysis of response assessment, correlation with PET/CT and clone evolution. Hematol Transfus Cell Ther 2024; 46 Suppl 6:S241-S249. [PMID: 39317576 PMCID: PMC11726095 DOI: 10.1016/j.htct.2024.07.005] [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: 05/29/2024] [Revised: 06/19/2024] [Accepted: 07/22/2024] [Indexed: 09/26/2024] Open
Abstract
INTRODUCTION Circulating tumor DNA (ctDNA) can be obtained from cell-free DNA (cfDNA) andis a new technique for genotyping, response assessment and prognosis in lymphoma. METHODS Eighteen patients with samples at diagnosis (ctDNA1), after treatment (ctDNA2) and extracted from diagnostic tissue (FFPE) were evaluated. RESULTS In all patients, at least one mutation in cfDNA was detected at diagnosis. CREBBP was the most frequent mutated gene (67 %). In 12 of the 15 patients with complete remission, the mutation attributed to the disease found at diagnosis cleared with treatment. A reduction in the ctDNA was observed after treatment in 14 patients, 12 of whom achieved complete remission. Correlations were found between the ctDNA at diagnosis and total metabolic tumor volume (r = 0.51; p-value = 0.014) and total lesion glycolysis 2.5 (r = 0.47; p-value = 0.024) by PET at diagnosis and between ctDNA at diagnosis and radiomic features of the lesions with the largest standardized uptake value. There was a strong inverse correlation between ΔctDNA1 and ΔSUVmax by PET/CT (r = -0.8788; p-value = 0.002). CONCLUSION Analysis of ctDNA and PET/CT in large B-cell lymphoma are complementary data for evaluating tumor burden and tumor clearance after treatment. Analysis of radiomic data might help to identify tumor characteristics and their changes after treatment.
Collapse
Affiliation(s)
- Guilherme Duffles
- University of Campinas, Hematology and Hemotherapy Centre, Hematology, Unicamp, Campinas 13083-878, SP, Brazil; Rede Dor Sao Luiz, Sao Paulo 01401-002, SP, Brazil.
| | | | | | | | | | - Leandro Freitas
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, 13083-888, SP, Brazil
| | - Fernanda Niemann
- University of Campinas, Hematology and Hemotherapy Centre, Hematology, Unicamp, Campinas 13083-878, SP, Brazil
| | | | - Celso Darío Ramos
- Division of Nuclear Medicine, School of Medical Sciences, University of Campinas (UNICAMP), Campinas 13083-888, SP, Brazil
| | | | - Irene Lorand-Metze
- University of Campinas, Hematology and Hemotherapy Centre, Hematology, Unicamp, Campinas 13083-878, SP, Brazil
| |
Collapse
|
5
|
Wang S, Belemlilga D, Lei Y, Ganti AKP, Lin C, Asif S, Marasco JT, Oh K, Zhou S. Enhancing Survival Outcome Predictions in Metastatic Non-Small Cell Lung Cancer Through PET Radiomics Analysis. Cancers (Basel) 2024; 16:3731. [PMID: 39594686 PMCID: PMC11592397 DOI: 10.3390/cancers16223731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
(1) Background: Advanced-stage lung cancer poses significant management challenges. The goal of this study was to identify crucial clinical and PET radiomics features that enable prognostic stratification for predicting outcomes. (2) Methods: PET radiomics features of the primary lung lesions were extracted from 99 patients with stage IVB NSCLC, and the robustness of these PET radiomics features was evaluated against uncertainties stemming from extraction parameters and contour variation. We trained three survival risk models (clinical, radiomics, and a composite) through a penalized Cox model framework. We also created a Balanced Random Forest classification predictive model, using the selected features, to predict 1-year survival. (3) Results: We identified 367 common PET radiomics features that exhibited robustness to perturbations introduced by contour variation and extraction parameters. Our findings indicated that both the radiomics and the composite model outperformed the clinical model in stratifying the risk for survival with statistical significance. In predicting 1-year survival, the radiomics model and the composite model also achieved better predicting accuracies compared to the clinical model. (4) Conclusions: Robust PET radiomics analysis successfully facilitated the stratification of patient risk for survival outcomes and predicted 1-year survival in stage IVB NSCLC.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Darryl Belemlilga
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
- College of Arts and Sciences, University of Nebraska Omaha, Omaha, NE 68182, USA
| | - Yu Lei
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Apar Kishor P Ganti
- Division of Oncology and Hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System, Omaha, NE 68105, USA;
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Samia Asif
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68105, USA;
| | - Jacob T Marasco
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Kyuhak Oh
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| | - Sumin Zhou
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (D.B.); (Y.L.); (C.L.); (J.T.M.); (K.O.); (S.Z.)
| |
Collapse
|
6
|
Stefano A. Challenges and limitations in applying radiomics to PET imaging: Possible opportunities and avenues for research. Comput Biol Med 2024; 179:108827. [PMID: 38964244 DOI: 10.1016/j.compbiomed.2024.108827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/05/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
Radiomics, the high-throughput extraction of quantitative imaging features from medical images, holds immense potential for advancing precision medicine in oncology and beyond. While radiomics applied to positron emission tomography (PET) imaging offers unique insights into tumor biology and treatment response, it is imperative to elucidate the challenges and constraints inherent in this domain to facilitate their translation into clinical practice. This review examines the challenges and limitations of applying radiomics to PET imaging, synthesizing findings from the last five years (2019-2023) and highlights the significance of addressing these challenges to realize the full clinical potential of radiomics in oncology and molecular imaging. A comprehensive search was conducted across multiple electronic databases, including PubMed, Scopus, and Web of Science, using keywords relevant to radiomics issues in PET imaging. Only studies published in peer-reviewed journals were eligible for inclusion in this review. Although many studies have highlighted the potential of radiomics in predicting treatment response, assessing tumor heterogeneity, enabling risk stratification, and personalized therapy selection, various challenges regarding the practical implementation of the proposed models still need to be addressed. This review illustrates the challenges and limitations of radiomics in PET imaging across various cancer types, encompassing both phantom and clinical investigations. The analyzed studies highlight the importance of reproducible segmentation methods, standardized pre-processing and post-processing methodologies, and the need to create large multicenter studies registered in a centralized database to promote the continuous validation and clinical integration of radiomics into PET imaging.
Collapse
Affiliation(s)
- Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy.
| |
Collapse
|
7
|
Nakamoto T, Nawa K, Nishiyama K, Yoshida K, Saito D, Horiguchi M, Shinya Y, Ohta T, Ozaki S, Nozawa Y, Minamitani M, Imae T, Abe O, Yamashita H, Nakagawa K. Neurological prognosis prediction for cardiac arrest patients using quantitative imaging biomarkers from brain computed tomography. Phys Med 2024; 125:103425. [PMID: 39142029 DOI: 10.1016/j.ejmp.2024.103425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/08/2024] [Accepted: 06/29/2024] [Indexed: 08/16/2024] Open
Abstract
PURPOSE We aimed to predict the neurological prognosis of cardiac arrest (CA) patients using quantitative imaging biomarkers extracted from brain computed tomography images. METHODS We retrospectively enrolled 86 CA patients (good prognosis, 32; poor prognosis, 54) who were treated at three hospitals between 2017 and 2019. We then extracted 1131 quantitative imaging biomarkers from whole-brain and local volumes of interest in the computed tomography images of the patients. The data were split into training and test sets containing 60 and 26 samples, respectively, and the training set was used to select representative quantitative imaging biomarkers for classification. In univariate analysis, the classification was evaluated using the p-value of the Brunner-Munzel test and area under the receiver operating characteristic curve (AUC) for the test set. In multivariate analysis, machine learning models reflecting nonlinear and complex relations were trained, and they were evaluated using the AUC on the test set. RESULTS The best performance provided p = 0.009 (<0.01) and an AUC of 0.775 (95% confidence interval, 0.590-0.960) for the univariate analysis and an AUCof0.813 (95% confidence interval, 0.640-0.985) for the multivariate analysis. Overall, the gray level with the maximum gradient in the histogram of the three-dimensionally low-pass-filtered image was an important feature for prediction across the analyses. CONCLUSIONS Quantitative imaging biomarkers can be used in neurological prognosis prediction for CA patients. Relevant biomarkers may contribute to protocolized computed tomography image acquisition to ensure proper decision support in acute care.
Collapse
Affiliation(s)
- Takahiro Nakamoto
- Department of Biological Science and Engineering, Faculty of Health Sciences, Hokkaido University, N12-W5, Kita-ku, Sapporo, Hokkaido 060-0812, Japan; Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Kanabu Nawa
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Kei Nishiyama
- Department of Emergency and Critical Care, Niigata University, 1-754 Asahimachidori, Chuo-ku, Niigata 951-8510, Japan
| | - Kosuke Yoshida
- Department of Emergency and Critical Care Medicine, National Hospital Organization Kyoto Medical Center, 1-1 Fukakusamukaihatacho, Fushimi-ku, Kyoto 612-8555, Japan
| | - Daizo Saito
- Division of Traumatology, National Defense Medical College Research Institute, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan
| | - Masahito Horiguchi
- Department of Emergency and Critical Care Medicine, Japanese Red Cross Kyoto Daiichi Hospital, 15-749 Honmachi, Higashiyama-ku, Kyoto 605-0981, Japan
| | - Yuki Shinya
- Department of Neurosurgery, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Takeshi Ohta
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Sho Ozaki
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan; Graduate School of Science and Technology, Hirosaki University, 3 Bunkyo, Hirosaki, Aomori 036-8561, Japan
| | - Yuki Nozawa
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masanari Minamitani
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Toshikazu Imae
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Osamu Abe
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hideomi Yamashita
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Keiichi Nakagawa
- Department of Radiology, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
8
|
Yang J, Xiao L, Zhou M, Li Y, Cai Y, Gan Y, Tang Y, Hu S. [ 68Ga]Ga‑PSMA‑617 PET-based radiomics model to identify candidates for active surveillance amongst patients with GGG 1-2 prostate cancer at biopsy. Cancer Imaging 2024; 24:86. [PMID: 38965552 PMCID: PMC11229016 DOI: 10.1186/s40644-024-00735-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024] Open
Abstract
PURPOSE To develop a radiomics-based model using [68Ga]Ga-PSMA PET/CT to predict postoperative adverse pathology (AP) in patients with biopsy Gleason Grade Group (GGG) 1-2 prostate cancer (PCa), assisting in the selection of patients for active surveillance (AS). METHODS A total of 75 men with biopsy GGG 1-2 PCa who underwent radical prostatectomy (RP) were enrolled. The patients were randomly divided into a training group (70%) and a testing group (30%). Radiomics features of entire prostate were extracted from the [68Ga]Ga-PSMA PET scans and selected using the minimum redundancy maximum relevance algorithm and the least absolute shrinkage and selection operator regression model. Logistic regression analyses were conducted to construct the prediction models. Receiver operating characteristic (ROC) curve, decision curve analysis (DCA), and calibration curve were employed to evaluate the diagnostic value, clinical utility, and predictive accuracy of the models, respectively. RESULTS Among the 75 patients, 30 had AP confirmed by RP. The clinical model showed an area under the curve (AUC) of 0.821 (0.695-0.947) in the training set and 0.795 (0.603-0.987) in the testing set. The radiomics model achieved AUC values of 0.830 (0.720-0.941) in the training set and 0.829 (0.624-1.000) in the testing set. The combined model, which incorporated the Radiomics score (Radscore) and free prostate-specific antigen (FPSA)/total prostate-specific antigen (TPSA), demonstrated higher diagnostic efficacy than both the clinical and radiomics models, with AUC values of 0.875 (0.780-0.970) in the training set and 0.872 (0.678-1.000) in the testing set. DCA showed that the net benefits of the combined model and radiomics model exceeded those of the clinical model. CONCLUSION The combined model shows potential in stratifying men with biopsy GGG 1-2 PCa based on the presence of AP at final pathology and outperforms models based solely on clinical or radiomics features. It may be expected to aid urologists in better selecting suitable patients for AS.
Collapse
Affiliation(s)
- Jinhui Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Yujia Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
| | - Yi Cai
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Gan
- Department of Urology, Disorders of Prostate Cancer Multidisciplinary Team, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, Bern, Switzerland.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders (XIANGYA), Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Key Laboratory of Biological, Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Song Y, Tian Y, Lu X, Chen G, Lv X. Prognostic value of 18F-FDG PET radiomics and sarcopenia in patients with oral squamous cell carcinoma. Med Phys 2024; 51:4907-4921. [PMID: 38252704 DOI: 10.1002/mp.16949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 11/28/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Oral cancer is one of the most common malignancies in the head and neck region. Approximately 90% of oral cancers are oral squamous cell carcinomas (OSCC). 18F-FDG PET/CT has been used in OSCC patients for its high value in detecting metastatic lymph nodes and distant metastases. PET radiomics and sarcopenia can be measured on the PET and CT components of 18F-FDG PET/CT. PURPOSE This study aimed to investigate the prognostic value of radiomics and sarcopenia measured on the PET and CT components of pre-operation 18F-FDG PET/CT in OSCC. METHODS A total of 116 patients eventually enrolled in our study were randomly divided into two cohorts: training cohort (n = 58) and validation cohort (n = 58). The Cox model combined with the least absolute shrinkage and selection operator (LASSO) algorithm was applied to construct the radiomics score (Rad_score). The third lumber skeletal muscle index (L3 SMI) was calculated to identify sarcopenia. Univariate and multivariate Cox regression analyses were performed to identify the independent prognostic factors. Based on the clinical factors, the clinical model was constructed, and the combined model was developed through the combination of the clinical model and Rad_score. C index, time-dependent C-index curves, receiver operating characteristic (ROC) curve, calibration curves, and decision curve analysis were used to evaluate the performance of prediction models. RESULTS Three radiomics features constitute the Rad_score for overall survival (OS) and progression-free survival (PFS), respectively. Multivariate Cox regression analysis revealed that Rad_score was an independent prognostic factor, whereas sarcopenia was not. The combined models showed satisfactory performance in both the training cohort (C-index: OS:0.836, PFS:0.776) and the validation cohort (C-index: OS:0.744, PFS:0.712). The combined models were visualized as nomograms. Nomogram scores can realize the risk stratification of OSCC patients. Lower nomogram score is significantly related to the poorer OS (training cohort: p < 0.0001, validation cohort: p < 0.0001, overall cohort: p < 0.0001) and PFS (training cohort: p < 0.0001, validation cohort: p = 0.00017, overall cohort: p < 0.0001). CONCLUSIONS Rad_score, but not sarcopenia, was an independent prognostic factor for patients with OSCC. The nomograms had a satisfactory performance, which might be helpful for OSCC patients and clinicians in personalized prognostic prediction and treatment decision-making.
Collapse
Affiliation(s)
- Yuxing Song
- Department of Oral & Maxillofacial Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Tian
- NanFang PET Center, Southern Medical University NanFang Hospital, Guangzhou, China
| | - Xinyan Lu
- Department of Oral & Maxillofacial Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gaoxiang Chen
- Department of Oral & Maxillofacial Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaozhi Lv
- Department of Oral & Maxillofacial Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Wang Y, Yang G, Gao X, Li L, Zhu H, Yi H. Subregion-specific 18F-FDG PET-CT radiomics for the pre-treatment prediction of EGFR mutation status in solid lung adenocarcinoma. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:134-143. [PMID: 38737644 PMCID: PMC11087292 DOI: 10.62347/ddrr4923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/07/2024] [Indexed: 05/14/2024]
Abstract
This study aimed to assess the efficacy of fluor-18 fluorodeoxyglucose (18F-FDG) PET/CT using sub-regional-based radiomics in predicting epidermal growth factor receptor (EGFR) mutation status in pretreatment patients with solid lung adenocarcinoma. A retrospective analysis included 269 patients (134 EGFR+ and 135 EGFR-) who underwent pretreatment 18F-FDG PET/CT scans and EGFR mutation testing. The most metabolically active intratumoral sub-region was identified, and radiomics features from whole tumors or sub-regional regions were used to build classification models. The dataset was split into a 7:3 ratio for training and independent testing. Feature subsets were determined by Pearson correlation and the Kruskal Wallis test and radiomics classifiers were built with support vector machines or logistic regressions. Evaluation metrics, including accuracy, area under the curve (AUC), sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) were employed for different classifiers. Results indicated that the sub-region-based classifier outperformed the whole-tumor classifier in terms of accuracy (73.8% vs. 66.2%), AUC (0.768 vs. 0.632), specificity (65.0% vs. 50.0%), PPV (70.2% vs. 62.2%), and NPV (78.8% vs. 74.0%). The clinical classifier exhibited an accuracy of 75.0%, AUC of 0.768, sensitivity of 72.5%, specificity of 77.5%, PPV of 76.3%, and NPV of 73.8%. The combined classifier, incorporating sub-region analysis and clinical parameters, demonstrated further improvement with an accuracy of 77.5%, AUC of 0.807, sensitivity of 77.5%, specificity of 77.5%, and NPV of 77.5%. The study suggests that sub-region-based 18F-FDG PET/CT radiomics enhances EGFR mutation prediction in solid lung adenocarcinoma, providing a practical and cost-efficient alternative to invasive EGFR testing.
Collapse
Affiliation(s)
- Yun Wang
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Guang Yang
- Department of Physics, Shanghai Key Laboratory of Magnetic Resonance, East China Normal UniversityShanghai 200062, China
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Linfa Li
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Hongzhou Zhu
- Department of Radiology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| | - Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, China
| |
Collapse
|
11
|
Sujit SJ, Aminu M, Karpinets TV, Chen P, Saad MB, Salehjahromi M, Boom JD, Qayati M, George JM, Allen H, Antonoff MB, Hong L, Hu X, Heeke S, Tran HT, Le X, Elamin YY, Altan M, Vokes NI, Sheshadri A, Lin J, Zhang J, Lu Y, Behrens C, Godoy MCB, Wu CC, Chang JY, Chung C, Jaffray DA, Wistuba II, Lee JJ, Vaporciyan AA, Gibbons DL, Heymach J, Zhang J, Cascone T, Wu J. Enhancing NSCLC recurrence prediction with PET/CT habitat imaging, ctDNA, and integrative radiogenomics-blood insights. Nat Commun 2024; 15:3152. [PMID: 38605064 PMCID: PMC11009351 DOI: 10.1038/s41467-024-47512-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
While we recognize the prognostic importance of clinicopathological measures and circulating tumor DNA (ctDNA), the independent contribution of quantitative image markers to prognosis in non-small cell lung cancer (NSCLC) remains underexplored. In our multi-institutional study of 394 NSCLC patients, we utilize pre-treatment computed tomography (CT) and 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) to establish a habitat imaging framework for assessing regional heterogeneity within individual tumors. This framework identifies three PET/CT subtypes, which maintain prognostic value after adjusting for clinicopathologic risk factors including tumor volume. Additionally, these subtypes complement ctDNA in predicting disease recurrence. Radiogenomics analysis unveil the molecular underpinnings of these imaging subtypes, highlighting downregulation in interferon alpha and gamma pathways in the high-risk subtype. In summary, our study demonstrates that these habitat imaging subtypes effectively stratify NSCLC patients based on their risk levels for disease recurrence after initial curative surgery or radiotherapy, providing valuable insights for personalized treatment approaches.
Collapse
Affiliation(s)
- Sheeba J Sujit
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Muhammad Aminu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tatiana V Karpinets
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pingjun Chen
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maliazurina B Saad
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Morteza Salehjahromi
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John D Boom
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mohamed Qayati
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James M George
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Haley Allen
- Natural Sciences, Rice University, Houston, TX, USA
| | - Mara B Antonoff
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lingzhi Hong
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Hu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simon Heeke
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hai T Tran
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiuning Le
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yasir Y Elamin
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie I Vokes
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ajay Sheshadri
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie Lin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yang Lu
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmen Behrens
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Myrna C B Godoy
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carol C Wu
- Department of Thoracic Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joe Y Chang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David A Jaffray
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Jack Lee
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ara A Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John Heymach
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Genomics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lung Cancer Interception Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tina Cascone
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jia Wu
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Institute of Data Science in Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Tapper W, Carneiro G, Mikropoulos C, Thomas SA, Evans PM, Boussios S. The Application of Radiomics and AI to Molecular Imaging for Prostate Cancer. J Pers Med 2024; 14:287. [PMID: 38541029 PMCID: PMC10971024 DOI: 10.3390/jpm14030287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/23/2024] [Accepted: 03/06/2024] [Indexed: 11/11/2024] Open
Abstract
Molecular imaging is a key tool in the diagnosis and treatment of prostate cancer (PCa). Magnetic Resonance (MR) plays a major role in this respect with nuclear medicine imaging, particularly, Prostate-Specific Membrane Antigen-based, (PSMA-based) positron emission tomography with computed tomography (PET/CT) also playing a major role of rapidly increasing importance. Another key technology finding growing application across medicine and specifically in molecular imaging is the use of machine learning (ML) and artificial intelligence (AI). Several authoritative reviews are available of the role of MR-based molecular imaging with a sparsity of reviews of the role of PET/CT. This review will focus on the use of AI for molecular imaging for PCa. It will aim to achieve two goals: firstly, to give the reader an introduction to the AI technologies available, and secondly, to provide an overview of AI applied to PET/CT in PCa. The clinical applications include diagnosis, staging, target volume definition for treatment planning, outcome prediction and outcome monitoring. ML and AL techniques discussed include radiomics, convolutional neural networks (CNN), generative adversarial networks (GAN) and training methods: supervised, unsupervised and semi-supervised learning.
Collapse
Affiliation(s)
- William Tapper
- Centre for Vision Speech and Signal Processing, The University of Surrey, 388 Stag Hill, Surrey, Guildford GU2 7XH, UK; (W.T.); (G.C.); (P.M.E.)
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, UK;
| | - Gustavo Carneiro
- Centre for Vision Speech and Signal Processing, The University of Surrey, 388 Stag Hill, Surrey, Guildford GU2 7XH, UK; (W.T.); (G.C.); (P.M.E.)
| | - Christos Mikropoulos
- Clinical Oncology, Royal Surrey NHS Foundation Trust, Egerton Road, Surrey, Guildford GU2 7XX, UK;
| | - Spencer A. Thomas
- National Physical Laboratory, Hampton Road, Teddington TW11 0LW, UK;
| | - Philip M. Evans
- Centre for Vision Speech and Signal Processing, The University of Surrey, 388 Stag Hill, Surrey, Guildford GU2 7XH, UK; (W.T.); (G.C.); (P.M.E.)
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham ME7 5NY, UK
- School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, Strand, London WC2R 2LS, UK
- Kent and Medway Medical School, University of Kent, Canterbury CT2 7LX, UK
- Faculty of Medicine, Health, and Social Care, Canterbury Christ Church University, Canterbury CT2 7PB, UK
- AELIA Organisation, 9th km Thessaloniki–Thermi, 57001 Thessaloniki, Greece
| |
Collapse
|
13
|
Yu L, Zhang Z, Yi H, Wang J, Li J, Wang X, Bai H, Ge H, Zheng X, Ni J, Qi H, Guan Y, Xu W, Zhu Z, Xing L, Dekker A, Wee L, Traverso A, Ye Z, Yuan Z. A PET/CT radiomics model for predicting distant metastasis in early-stage non-small cell lung cancer patients treated with stereotactic body radiotherapy: a multicentric study. Radiat Oncol 2024; 19:10. [PMID: 38254106 PMCID: PMC10802016 DOI: 10.1186/s13014-024-02402-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVES Stereotactic body radiotherapy (SBRT) is a treatment option for patients with early-stage non-small cell lung cancer (NSCLC) who are unfit for surgery. Some patients may experience distant metastasis. This study aimed to develop and validate a radiomics model for predicting distant metastasis in patients with early-stage NSCLC treated with SBRT. METHODS Patients at five institutions were enrolled in this study. Radiomics features were extracted based on the PET/CT images. After feature selection in the training set (from Tianjin), CT-based and PET-based radiomics signatures were built. Models based on CT and PET signatures were built and validated using external datasets (from Zhejiang, Zhengzhou, Shandong, and Shanghai). An integrated model that included CT and PET radiomic signatures was developed. The performance of the proposed model was evaluated in terms of its discrimination, calibration, and clinical utility. Multivariate logistic regression was used to calculate the probability of distant metastases. The cutoff value was obtained using the receiver operator characteristic curve (ROC), and the patients were divided into high- and low-risk groups. Kaplan-Meier analysis was used to evaluate the distant metastasis-free survival (DMFS) of different risk groups. RESULTS In total, 228 patients were enrolled. The median follow-up time was 31.4 (2.0-111.4) months. The model based on CT radiomics signatures had an area under the curve (AUC) of 0.819 in the training set (n = 139) and 0.786 in the external dataset (n = 89). The PET radiomics model had an AUC of 0.763 for the training set and 0.804 for the external dataset. The model combining CT and PET radiomics had an AUC of 0.835 for the training set and 0.819 for the external dataset. The combined model showed a moderate calibration and a positive net benefit. When the probability of distant metastasis was greater than 0.19, the patient was considered to be at high risk. The DMFS of patients with high- and low-risk was significantly stratified (P < 0.001). CONCLUSIONS The proposed PET/CT radiomics model can be used to predict distant metastasis in patients with early-stage NSCLC treated with SBRT and provide a reference for clinical decision-making. In this study, the model was established by combining CT and PET radiomics signatures in a moderate-quantity training cohort of early-stage NSCLC patients treated with SBRT and was successfully validated in independent cohorts. Physicians could use this easy-to-use model to assess the risk of distant metastasis after SBRT. Identifying subgroups of patients with different risk factors for distant metastasis is useful for guiding personalized treatment approaches.
Collapse
Affiliation(s)
- Lu Yu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhen Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - HeQing Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jin Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Junyi Li
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Xiaofeng Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Hui Bai
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Hong Ge
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoli Zheng
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haoran Qi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Yong Guan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Wengui Xu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Zhaoxiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
| | - Zhiyong Yuan
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
14
|
Zhang M, Wang Y, Lv M, Sang L, Wang X, Yu Z, Yang Z, Wang Z, Sang L. Trends and Hotspots in Global Radiomics Research: A Bibliometric Analysis. Technol Cancer Res Treat 2024; 23:15330338241235769. [PMID: 38465611 DOI: 10.1177/15330338241235769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Objectives: The purpose of this research is to summarize the structure of radiomics-based knowledge and to explore potential trends and priorities by using bibliometric analysis. Methods: Select radiomics-related publications from 2012 to October 2022 from the Science Core Collection Web site. Use VOSviewer (version 1.6.18), CiteSpace (version 6.1.3), Tableau (version 2022), Microsoft Excel and Rstudio's free online platforms (http://bibliometric.com) for co-writing, co-citing, and co-occurrence analysis of countries, institutions, authors, references, and keywords in the field. The visual analysis is also carried out on it. Results: The study included 6428 articles. Since 2012, there has been an increase in research papers based on radiomics. Judging by publications, China has made the largest contribution in this area. We identify the most productive institutions and authors as Fudan University and Tianjie. The top three magazines with the most publications are《FRONTIERS IN ONCOLOGY》, 《EUROPEAN RADIOLOGY》, and 《CANCERS》. According to the results of reference and keyword analysis, "deep learning, nomogram, ultrasound, f-18-fdg, machine learning, covid-19, radiogenomics" has been determined as the main research direction in the future. Conclusion: Radiomics is in a phase of vigorous development with broad prospects. Cross-border cooperation between countries and institutions should be strengthened in the future. It can be predicted that the development of deep learning-based models and multimodal fusion models will be the focus of future research. Advances in knowledge: This study explores the current state of research and hot spots in the field of radiomics from multiple perspectives, comprehensively, and objectively reflecting the evolving trends in imaging-related research and providing a reference for future research.
Collapse
Affiliation(s)
- Minghui Zhang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Yan Wang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Mutian Lv
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Li Sang
- Department of Acupuncture and Massage, Shouguang Hospital of Traditional Chinese Medicine, Weifang, P. R. China
| | - Xuemei Wang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zijun Yu
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Ziyi Yang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Zhongqing Wang
- Department of Information Center, The First Hospital of China Medical University, Shenyang, P. R. China
| | - Liang Sang
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
15
|
Williams TL, Gonen M, Wray R, Do RKG, Simpson AL. Quantitation of Oncologic Image Features for Radiomic Analyses in PET. Methods Mol Biol 2024; 2729:409-421. [PMID: 38006509 DOI: 10.1007/978-1-0716-3499-8_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Radiomics is an emerging and exciting field of study involving the extraction of many quantitative features from radiographic images. Positron emission tomography (PET) images are used in cancer diagnosis and staging. Utilizing radiomics on PET images can better quantify the spatial relationships between image voxels and generate more consistent and accurate results for diagnosis, prognosis, treatment, etc. This chapter gives the general steps a researcher would take to extract PET radiomic features from medical images and properly develop models to implement.
Collapse
Affiliation(s)
- Travis L Williams
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gonen
- Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rick Wray
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard K G Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amber L Simpson
- School of Computing and Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
16
|
Rahmim A, Toosi A, Salmanpour MR, Dubljevic N, Janzen I, Shiri I, Yuan R, Ho C, Zaidi H, MacAulay C, Uribe C, Yousefirizi F. Tensor radiomics: paradigm for systematic incorporation of multi-flavoured radiomics features. Quant Imaging Med Surg 2023; 13:7680-7694. [PMID: 38106259 PMCID: PMC10722050 DOI: 10.21037/qims-23-163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/02/2023] [Indexed: 12/19/2023]
Abstract
Background Radiomics features hold significant value as quantitative imaging biomarkers for diagnosis, prognosis, and treatment response assessment. To generate radiomics features and ultimately develop signatures, various factors can be manipulated, including image discretization parameters (e.g., bin number or size), convolutional filters, segmentation perturbation, or multi-modality fusion levels. Typically, only one set of parameters is employed, resulting in a single value or "flavour" for each radiomics feature. In contrast, we propose "tensor radiomics" (TR) where tensors of features calculated using multiple parameter combinations (i.e., flavours) are utilized to optimize the creation of radiomics signatures. Methods We provide illustrative instances of TR implementation in positron emission tomography-computed tomography (PET-CT), magnetic resonance imaging (MRI), and CT by leveraging machine learning (ML) and deep learning (DL) methodologies, as well as reproducibility analyses: (I) to predict overall survival (OS) in lung cancer (CT) and head and neck cancer (PET-CT), TR was employed by varying bin sizes. This approach involved use of a hybrid deep neural network called 'TR-Net' and two ML-based techniques for combining different flavours. (II) TR was constructed by incorporating different segmentation perturbations and various bin sizes to classify the response of late-stage lung cancer to first-line immunotherapy using CT images. (III) In MRI of glioblastoma (GBM), TR was implemented to generate multi-flavour radiomics features, enabling enhanced analysis and interpretation. (IV) TR was employed via multiple PET-CT fusions in head and neck cancer. Flavours based on different fusions were created using Laplacian pyramids and wavelet transforms. Results Our findings demonstrated that TR outperformed conventional radiomics features in lung cancer CT and head and neck cancer PET-CT images, significantly enhancing OS prediction accuracy. TR also improved classification of lung cancer response to therapy and exhibited notable advantages in reproducibility compared to single-flavour features in MR imaging of GBM. Moreover, in head and neck cancer, TR through multiple PET-CT fusions exhibited improved performance in predicting OS. Conclusions We conclude that the proposed TR paradigm has significant potential to improve performance in different medical imaging tasks. By incorporating multiple flavours of radiomics features, TR overcomes limitations associated with individual features and shows promise in enhancing prognostic capabilities in clinical settings.
Collapse
Affiliation(s)
- Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada
| | - Amirhosein Toosi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | | | - Natalia Dubljevic
- Department of Physics & Astronomy, University of British Columbia, Vancouver, Canada
| | - Ian Janzen
- BC Cancer Research Institute, Vancouver, Canada
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Ren Yuan
- BC Cancer Research Institute, Vancouver, Canada
| | - Cheryl Ho
- BC Cancer Research Institute, Vancouver, Canada
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | | | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | | |
Collapse
|
17
|
Xie J, Xue B, Bian S, Ji X, Lin J, Zheng X, Tang K. A radiomics nomogram based on 18 F-FDG PET/CT and clinical risk factors for the prediction of peritoneal metastasis in gastric cancer. Nucl Med Commun 2023; 44:977-987. [PMID: 37578301 DOI: 10.1097/mnm.0000000000001742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
PURPOSE Peritoneal metastasis (PM) is usually considered an incurable factor of gastric cancer (GC) and not fit for surgery. The aim of this study is to develop and validate an 18 F-FDG PET/CT-derived radiomics model combining with clinical risk factors for predicting PM of GC. METHOD In this retrospective study, 410 GC patients (PM - = 281, PM + = 129) who underwent preoperative 18 F-FDG PET/CT images from January 2015 to October 2021 were analyzed. The patients were randomly divided into a training cohort (n = 288) and a validation cohort (n = 122). The maximum relevance and minimum redundancy (mRMR) and the least shrinkage and selection operator method were applied to select feature. Multivariable logistic regression analysis was preformed to develop the predicting model. Discrimination, calibration, and clinical usefulness were used to evaluate the performance of the nomogram. RESULT Fourteen radiomics feature parameters were selected to construct radiomics model. The area under the curve (AUC) of the radiomics model were 0.86 [95% confidence interval (CI), 0.81-0.90] in the training cohort and 0.85 (95% CI, 0.78-0.92) in the validation cohort. After multivariable logistic regression, peritoneal effusion, mean standardized uptake value (SUVmean), carbohydrate antigen 125 (CA125) and radiomics signature showed statistically significant differences between different PM status patients( P < 0.05). They were chosen to construct the comprehensive predicting model which showed a performance with an AUC of 0.92 (95% CI, 0.89-0.95) in the training cohort and 0.92 (95% CI, 0.86-0.98) in the validation cohort, respectively. CONCLUSION The nomogram based on 18 F-FDG PET/CT radiomics features and clinical risk factors can be potentially applied in individualized treatment strategy-making for GC patients before the surgery.
Collapse
Affiliation(s)
- Jiageng Xie
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Beihui Xue
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuying Bian
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaowei Ji
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jie Lin
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangwu Zheng
- Departments of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kun Tang
- Departments of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
18
|
Li Y, Han D, Shen C, Duan X. Construction of a comprehensive predictive model for axillary lymph node metastasis in breast cancer: a retrospective study. BMC Cancer 2023; 23:1028. [PMID: 37875818 PMCID: PMC10594862 DOI: 10.1186/s12885-023-11498-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023] Open
Abstract
PURPOSE The accurate assessment of axillary lymph node metastasis (LNM) in early-stage breast cancer (BC) is of great importance. This study aimed to construct an integrated model based on clinicopathology, ultrasound, PET/CT, and PET radiomics for predicting axillary LNM in early stage of BC. MATERIALS AND METHODS 124 BC patients who underwent 18 F-fluorodeoxyglucose (18 F-FDG) PET/CT and whose diagnosis were confirmed by surgical pathology were retrospectively analyzed and included in this study. Ultrasound, PET and clinicopathological features of all patients were analyzed, and PET radiomics features were extracted to establish an ultrasound model (clinicopathology and ultrasound; model 1), a PET model (clinicopathology, ultrasound, and PET; model 2), and a comprehensive model (clinicopathology, ultrasound, PET, and radiomics; model 3), and the diagnostic efficacy of each model was evaluated and compared. RESULTS The T stage, US_BIRADS, US_LNM, and PET_LNM in the positive axillary LNM group was significantly higher than that of in the negative LNM group (P = 0.013, P = 0.049, P < 0.001, P < 0.001, respectively). Radiomics score for predicting LNM (RS_LNM) for the negative LNM and positive LNM were statistically significant difference (-1.090 ± 0.448 vs. -0.693 ± 0.344, t = -4.720, P < 0.001), and the AUC was 0.767 (95% CI: 0.674-0.861). The ROC curves showed that model 3 outperformed model 1 for the sensitivity (model 3 vs. model 1, 82.86% vs. 48.57%), and outperformed model 2 for the specificity (model 3 vs. model 2, 82.02% vs. 68.54%) in the prediction of LNM. The AUC of mode 1, model 2 and model 3 was 0.687, 0.826 and 0.874, and the Delong test showed the AUC of model 3 was significantly higher than that of model 1 and model 2 (P < 0.05). Decision curve analysis showed that model 3 resulted in a higher degree of net benefit for all the patients than model 1 and model 2. CONCLUSION The use of a comprehensive model based on clinicopathology, ultrasound, PET/CT, and PET radiomics can effectively improve the diagnostic efficacy of axillary LNM in BC. TRIAL REGISTRATION This study was registered at ClinicalTrials Gov (number NCT05826197) on 7th, May 2023.
Collapse
Affiliation(s)
- Yan Li
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an Shaanxi, 710061, China
| | - Dong Han
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an Shaanxi, 710061, China
| | - Cong Shen
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an Shaanxi, 710061, China
| | - Xiaoyi Duan
- PET/CT Center, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an Shaanxi, 710061, China.
| |
Collapse
|
19
|
Balma M, Laudicella R, Gallio E, Gusella S, Lorenzon L, Peano S, Costa RP, Rampado O, Farsad M, Evangelista L, Deandreis D, Papaleo A, Liberini V. Applications of Artificial Intelligence and Radiomics in Molecular Hybrid Imaging and Theragnostics for Neuro-Endocrine Neoplasms (NENs). Life (Basel) 2023; 13:1647. [PMID: 37629503 PMCID: PMC10455722 DOI: 10.3390/life13081647] [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: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Nuclear medicine has acquired a crucial role in the management of patients with neuroendocrine neoplasms (NENs) by improving the accuracy of diagnosis and staging as well as their risk stratification and personalized therapies, including radioligand therapies (RLT). Artificial intelligence (AI) and radiomics can enable physicians to further improve the overall efficiency and accuracy of the use of these tools in both diagnostic and therapeutic settings by improving the prediction of the tumor grade, differential diagnosis from other malignancies, assessment of tumor behavior and aggressiveness, and prediction of treatment response. This systematic review aims to describe the state-of-the-art AI and radiomics applications in the molecular imaging of NENs.
Collapse
Affiliation(s)
- Michele Balma
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy; (S.P.); (A.P.); (V.L.)
| | - Riccardo Laudicella
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.L.); (R.P.C.)
| | - Elena Gallio
- Medical Physics Unit, A.O.U. Città Della Salute E Della Scienza Di Torino, Corso Bramante 88/90, 10126 Torino, Italy; (E.G.); (O.R.)
| | - Sara Gusella
- Nuclear Medicine, Central Hospital Bolzano, 39100 Bolzano, Italy; (S.G.); (M.F.)
| | - Leda Lorenzon
- Medical Physics Department, Central Bolzano Hospital, 39100 Bolzano, Italy;
| | - Simona Peano
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy; (S.P.); (A.P.); (V.L.)
| | - Renato P. Costa
- Unit of Nuclear Medicine, Biomedical Department of Internal and Specialist Medicine, University of Palermo, 90133 Palermo, Italy; (R.L.); (R.P.C.)
| | - Osvaldo Rampado
- Medical Physics Unit, A.O.U. Città Della Salute E Della Scienza Di Torino, Corso Bramante 88/90, 10126 Torino, Italy; (E.G.); (O.R.)
| | - Mohsen Farsad
- Nuclear Medicine, Central Hospital Bolzano, 39100 Bolzano, Italy; (S.G.); (M.F.)
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, 20089 Milan, Italy;
| | - Desiree Deandreis
- Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy and Université Paris Saclay, 94805 Villejuif, France;
| | - Alberto Papaleo
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy; (S.P.); (A.P.); (V.L.)
| | - Virginia Liberini
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100 Cuneo, Italy; (S.P.); (A.P.); (V.L.)
| |
Collapse
|
20
|
Samimi R, Shiri I, Ahmadyar Y, van den Hoff J, Kamali-Asl A, Rezaee A, Yousefirizi F, Geramifar P, Rahmim A. Radiomics predictive modeling from dual-time-point FDG PET K i parametric maps: application to chemotherapy response in lymphoma. EJNMMI Res 2023; 13:70. [PMID: 37493872 PMCID: PMC10371962 DOI: 10.1186/s13550-023-01022-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/18/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND To investigate the use of dynamic radiomics features derived from dual-time-point (DTP-feature) [18F]FDG PET metabolic uptake rate Ki parametric maps to develop a predictive model for response to chemotherapy in lymphoma patients. METHODS We analyzed 126 lesions from 45 lymphoma patients (responding n = 75 and non-responding n = 51) treated with chemotherapy from two different centers. Static and DTP radiomics features were extracted from baseline static PET images and DTP Ki parametric maps. Spearman's rank correlations were calculated between static and DTP features to identify features with potential additional information. We first employed univariate analysis to determine correlations between individual features, and subsequently utilized multivariate analysis to derive predictive models utilizing DTP and static radiomics features before and after ComBat harmonization. For multivariate modeling, we utilized both the minimum redundancy maximum relevance feature selection technique and the XGBoost classifier. To evaluate our model, we partitioned the patient datasets into training/validation and testing sets using an 80/20% split. Different metrics for classification including area under the curve (AUC), sensitivity (SEN), specificity (SPE), and accuracy (ACC) were reported in test sets. RESULTS Via Spearman's rank correlations, there was negligible to moderate correlation between 32 out of 65 DTP features and some static features (ρ < 0.7); all the other 33 features showed high correlations (ρ ≥ 0.7). In univariate modeling, no significant difference between AUC of DTP and static features was observed. GLRLM_RLNU from static features demonstrated a strong correlation (AUC = 0.75, p value = 0.0001, q value = 0.0007) with therapy response. The most predictive DTP features were GLCM_Energy, GLCM_Entropy, and Uniformity, each with AUC = 0.73, p value = 0.0001, and q value < 0.0005. In multivariate analysis, the mean ranges of AUCs increased following harmonization. Use of harmonization plus combining DTP and static features was shown to provide significantly improved predictions (AUC = 0.97 ± 0.02, accuracy = 0.89 ± 0.05, sensitivity = 0.92 ± 0.09, and specificity = 0.88 ± 0.05). All models depicted significant performance in terms of AUC, ACC, SEN, and SPE (p < 0.05, Mann-Whitney test). CONCLUSIONS Our results demonstrate significant value in harmonization of radiomics features as well as combining DTP and static radiomics models for predicting response to chemotherapy in lymphoma patients.
Collapse
Affiliation(s)
- Rezvan Samimi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, 1211, Geneva 4, Switzerland
| | - Yashar Ahmadyar
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran
| | - Jörg van den Hoff
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328, Dresden, Germany
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307, Dresden, Germany
| | - Alireza Kamali-Asl
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran.
| | | | - Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Parham Geramifar
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
21
|
Barrington SF. Advances in positron emission tomography and radiomics. Hematol Oncol 2023; 41 Suppl 1:11-19. [PMID: 37294959 PMCID: PMC10775708 DOI: 10.1002/hon.3137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 06/11/2023]
Abstract
Positron emission tomography is established for staging and response evaluation in lymphoma using visual evaluation and semi-quantitative analysis. Radiomic analysis involving quantitative imaging features at baseline, such as metabolic tumor volume and markers of disease dissemination and changes in the standardized uptake value during treatment are emerging as powerful biomarkers. The combination of radiomic features with clinical risk factors and genomic analysis offers the potential to improve clinical risk prediction. This review discusses the state of current knowledge, progress toward standardization of tumor delineation for radiomic analysis and argues that radiomic features, molecular markers and circulating tumor DNA should be included in clinical trial designs to enable the development of baseline and dynamic risk scores that could further advance the field to facilitate testing of novel treatments and personalized therapy in aggressive lymphomas.
Collapse
Affiliation(s)
- Sally F. Barrington
- School of Biomedical Engineering and Imaging SciencesSt Thomas' Campus, Kings College LondonLondonUK
| |
Collapse
|
22
|
Chan TH, Haworth A, Wang A, Osanlouy M, Williams S, Mitchell C, Hofman MS, Hicks RJ, Murphy DG, Reynolds HM. Detecting localised prostate cancer using radiomic features in PSMA PET and multiparametric MRI for biologically targeted radiation therapy. EJNMMI Res 2023; 13:34. [PMID: 37099047 PMCID: PMC10133419 DOI: 10.1186/s13550-023-00984-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 04/27/2023] Open
Abstract
BACKGROUND Prostate-Specific Membrane Antigen (PSMA) PET/CT and multiparametric MRI (mpMRI) are well-established modalities for identifying intra-prostatic lesions (IPLs) in localised prostate cancer. This study aimed to investigate the use of PSMA PET/CT and mpMRI for biologically targeted radiation therapy treatment planning by: (1) analysing the relationship between imaging parameters at a voxel-wise level and (2) assessing the performance of radiomic-based machine learning models to predict tumour location and grade. METHODS PSMA PET/CT and mpMRI data from 19 prostate cancer patients were co-registered with whole-mount histopathology using an established registration framework. Apparent Diffusion Coefficient (ADC) maps were computed from DWI and semi-quantitative and quantitative parameters from DCE MRI. Voxel-wise correlation analysis was conducted between mpMRI parameters and PET Standardised Uptake Value (SUV) for all tumour voxels. Classification models were built using radiomic and clinical features to predict IPLs at a voxel level and then classified further into high-grade or low-grade voxels. RESULTS Perfusion parameters from DCE MRI were more highly correlated with PET SUV than ADC or T2w. IPLs were best detected with a Random Forest Classifier using radiomic features from PET and mpMRI rather than either modality alone (sensitivity, specificity and area under the curve of 0.842, 0.804 and 0.890, respectively). The tumour grading model had an overall accuracy ranging from 0.671 to 0.992. CONCLUSIONS Machine learning classifiers using radiomic features from PSMA PET and mpMRI show promise for predicting IPLs and differentiating between high-grade and low-grade disease, which could be used to inform biologically targeted radiation therapy planning.
Collapse
Affiliation(s)
- Tsz Him Chan
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Annette Haworth
- Institute of Medical Physics, School of Physics, The University of Sydney, Sydney, NSW, Australia
| | - Alan Wang
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
- Centre for Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, The University of Auckland, Auckland, New Zealand
| | - Mahyar Osanlouy
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Scott Williams
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Rodney J Hicks
- Department of Medicine, St Vincent's Hospital Medical School, The University of Melbourne, Melbourne, VIC, Australia
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Hayley M Reynolds
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
23
|
Fukai S, Daisaki H, Ishiyama M, Shimada N, Umeda T, Motegi K, Ito R, Terauchi T. Reproducibility of the principal component analysis (PCA)-based data-driven respiratory gating on texture features in non-small cell lung cancer patients with 18 F-FDG PET/CT. J Appl Clin Med Phys 2023; 24:e13967. [PMID: 36943700 PMCID: PMC10161026 DOI: 10.1002/acm2.13967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/15/2023] [Accepted: 03/01/2023] [Indexed: 03/23/2023] Open
Abstract
OBJECTIVE Texture analysis is one of the lung cancer countermeasures in the field of radiomics. Even though image quality affects texture features, the reproducibility of principal component analysis (PCA)-based data-driven respiratory gating (DDG) on texture features remains poorly understood. Hence, this study aimed to clarify the reproducibility of PCA-based DDG on texture features in non-small cell lung cancer (NSCLC) patients with 18 F-Fluorodeoxyglucose (18 F-FDG) Positron emission tomography/computed tomography (PET/CT). METHODS Twenty patients with NSCLC who underwent 18 F-FDG PET/CT in routine clinical practice were retrospectively analyzed. Each patient's PET data were reconstructed in two PET groups of no gating (NG-PET) and PCA-based DDG gating (DDG-PET). Forty-six image features were analyzed using LIFEx software. Reproducibility was evaluated using Lin's concordance correlation coefficient ( ρ c ${\rho _c}$ ) and percentage difference (%Diff). Non-reproducibility was defined as having unacceptable strength ( ρ c $({\rho _c}$ < 0.8) and a %Diff of >10%. NG-PET and DDG-PET were compared using the Wilcoxon signed-rank test. RESULTS A total of 3/46 (6.5%) image features had unacceptable strength, and 9/46 (19.6%) image features had a %Diff of >10%. Significant differences between the NG-PET and DDG-PET groups were confirmed in only 4/46 (8.7%) of the high %Diff image features. CONCLUSION Although the DDG application affected several texture features, most image features had adequate reproducibility. PCA-based DDG-PET can be routinely used as interchangeable images for texture feature extraction from NSCLC patients.
Collapse
Affiliation(s)
- Shohei Fukai
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
- Graduate School of Radiological Technology, Gunma Prefectural College of Health Sciences, Gunma, Japan
| | - Hiromitsu Daisaki
- Graduate School of Radiological Technology, Gunma Prefectural College of Health Sciences, Gunma, Japan
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mitsutomi Ishiyama
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Naoki Shimada
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takuro Umeda
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kazuki Motegi
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryoma Ito
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Terauchi
- Department of Nuclear Medicine, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
24
|
Alberts I, Sari H, Mingels C, Afshar-Oromieh A, Pyka T, Shi K, Rominger A. Long-axial field-of-view PET/CT: perspectives and review of a revolutionary development in nuclear medicine based on clinical experience in over 7000 patients. Cancer Imaging 2023; 23:28. [PMID: 36934273 PMCID: PMC10024603 DOI: 10.1186/s40644-023-00540-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 02/25/2023] [Indexed: 03/20/2023] Open
Abstract
Recently introduced long-axial field-of-view (LAFOV) PET/CT systems represent one of the most significant advancements in nuclear medicine since the advent of multi-modality PET/CT imaging. The higher sensitivity exhibited by such systems allow for reductions in applied activity and short duration scans. However, we consider this to be just one small part of the story: Instead, the ability to image the body in its entirety in a single FOV affords insights which standard FOV systems cannot provide. For example, we now have the ability to capture a wider dynamic range of a tracer by imaging it over multiple half-lives without detrimental image noise, to leverage lower radiopharmaceutical doses by using dual-tracer techniques and with improved quantification. The potential for quantitative dynamic whole-body imaging using abbreviated protocols potentially makes these techniques viable for routine clinical use, transforming PET-reporting from a subjective analysis of semi-quantitative maps of radiopharmaceutical uptake at a single time-point to an accurate and quantitative, non-invasive tool to determine human function and physiology and to explore organ interactions and to perform whole-body systems analysis. This article will share the insights obtained from 2 years' of clinical operation of the first Biograph Vision Quadra (Siemens Healthineers) LAFOV system. It will also survey the current state-of-the-art in PET technology. Several technologies are poised to furnish systems with even greater sensitivity and resolution than current systems, potentially with orders of magnitude higher sensitivity. Current barriers which remain to be surmounted, such as data pipelines, patient throughput and the hindrances to implementing kinetic analysis for routine patient care will also be discussed.
Collapse
Affiliation(s)
- Ian Alberts
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Hasan Sari
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland
| | - Clemens Mingels
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Thomas Pyka
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Freiburgstr. 18, 3010, Bern, Switzerland.
| |
Collapse
|
25
|
Mansouri N, Balvay D, Zenteno O, Facchin C, Yoganathan T, Viel T, Herraiz JL, Tavitian B, Pérez-Liva M. Machine Learning of Multi-Modal Tumor Imaging Reveals Trajectories of Response to Precision Treatment. Cancers (Basel) 2023; 15:1751. [PMID: 36980637 PMCID: PMC10046832 DOI: 10.3390/cancers15061751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
The standard assessment of response to cancer treatments is based on gross tumor characteristics, such as tumor size or glycolysis, which provide very indirect information about the effect of precision treatments on the pharmacological targets of tumors. Several advanced imaging modalities allow for the visualization of targeted tumor hallmarks. Descriptors extracted from these images can help establishing new classifications of precision treatment response. We propose a machine learning (ML) framework to analyze metabolic-anatomical-vascular imaging features from positron emission tomography, ultrafast Doppler, and computed tomography in a mouse model of paraganglioma undergoing anti-angiogenic treatment with sunitinib. Imaging features from the follow-up of sunitinib-treated (n = 8, imaged once-per-week/6-weeks) and sham-treated (n = 8, imaged once-per-week/3-weeks) mice groups were dimensionally reduced and analyzed with hierarchical clustering Analysis (HCA). The classes extracted from HCA were used with 10 ML classifiers to find a generalized tumor stage prediction model, which was validated with an independent dataset of sunitinib-treated mice. HCA provided three stages of treatment response that were validated using the best-performing ML classifier. The Gaussian naive Bayes classifier showed the best performance, with a training accuracy of 98.7 and an average area under curve of 100. Our results show that metabolic-anatomical-vascular markers allow defining treatment response trajectories that reflect the efficacy of an anti-angiogenic drug on the tumor target hallmark.
Collapse
Affiliation(s)
| | - Daniel Balvay
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
| | - Omar Zenteno
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
| | - Caterina Facchin
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
- Cancer Drug Research Laboratory, Department of Medicine, Division of Medical Oncology, The Research Institute of the McGill University Health Center (RI-MUHC), Montréal, QC H4A 3J1, Canada
| | | | - Thomas Viel
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
| | - Joaquin Lopez Herraiz
- Nuclear Physics Group and IPARCOS, Department of Structure of Matter, Thermal Physics and Electronics, CEI Moncloa, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Bertrand Tavitian
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
- Radiology Department, AP-HP, European Hospital Georges Pompidou, F-75015 Paris, France
| | - Mailyn Pérez-Liva
- INSERM, PARCC, Université Paris Cité, F-75015 Paris, France
- Nuclear Physics Group and IPARCOS, Department of Structure of Matter, Thermal Physics and Electronics, CEI Moncloa, Universidad Complutense de Madrid, 28040 Madrid, Spain
| |
Collapse
|
26
|
Xu X, Sun X, Ma L, Zhang H, Ji W, Xia X, Lan X. 18F-FDG PET/CT radiomics signature and clinical parameters predict progression-free survival in breast cancer patients: A preliminary study. Front Oncol 2023; 13:1149791. [PMID: 36969043 PMCID: PMC10036789 DOI: 10.3389/fonc.2023.1149791] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 03/12/2023] Open
Abstract
IntroductionThis study aimed to investigate the feasibility of predicting progression-free survival (PFS) in breast cancer patients using pretreatment 18F-fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) radiomics signature and clinical parameters.MethodsBreast cancer patients who underwent 18F-FDG PET/CT imaging before treatment from January 2012 to December 2020 were eligible for study inclusion. Eighty-seven patients were randomly divided into training (n = 61) and internal test sets (n = 26) and an additional 25 patients were used as the external validation set. Clinical parameters, including age, tumor size, molecularsubtype, clinical TNM stage, and laboratory findings were collected. Radiomics features were extracted from preoperative PET/CT images. Least absolute shrinkage and selection operators were applied to shrink feature size and build a predictive radiomics signature. Univariate and multivariate Cox proportional hazards models and Kaplan-Meier analysis were used to assess the association of rad-score and clinical parameter with PFS. Nomograms were constructed to visualize survival prediction. C-index and calibration curve were used to evaluate nomogram performance.ResultsEleven radiomics features were selected to generate rad-score. The clinical model comprised three parameters: clinical M stage, CA125, and pathological N stage. Rad-score and clinical-model were significantly associated with PFS in the training set (P< 0.01) but not the test set. The integrated clinical-radiomics (ICR) model was significantly associated with PFS in both the training and test sets (P< 0.01). The ICR model nomogram had a significantly higher C-index than the clinical model and rad-score in the training and test sets. The C-index of the ICR model in the external validation set was 0.754 (95% confidence interval, 0.726–0.812). PFS significantly differed between the low- and high-risk groups stratified by the nomogram (P = 0.009). The calibration curve indicated the ICR model provided the greatest clinical benefit.ConclusionThe ICR model, which combined clinical parameters and preoperative 18F-FDG PET/CT imaging, was able to independently predict PFS in breast cancer patients and was superior to the clinical model alone and rad-score alone.
Collapse
Affiliation(s)
- Xiaojun Xu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, China
| | - Xun Sun
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, China
| | - Ling Ma
- He Kang Corporate Management (SH) Co. Ltd, Shanghai, China
| | - Huangqi Zhang
- Department of Radiology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenbin Ji
- Department of Radiology, Affiliated Taizhou Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Xiaotian Xia
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, China
- *Correspondence: Xiaotian Xia, ; Xiaoli Lan,
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
- Key Laboratory of Biological Targeted Therapy of the Ministry of Education, Wuhan, China
- *Correspondence: Xiaotian Xia, ; Xiaoli Lan,
| |
Collapse
|
27
|
Qian WL, Chen Q, Zhang JB, Xu JM, Hu CH. RESOLVE-based radiomics in cervical cancer: improved image quality means better feature reproducibility? Clin Radiol 2023; 78:e469-e476. [PMID: 37029000 DOI: 10.1016/j.crad.2023.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 04/07/2023]
Abstract
AIM To compare the reproducibility of apparent diffusion coefficient (ADC)-based radiomic features between readout-segmented echo-planar diffusion-weighted imaging (RESOLVE) and single-shot echo-planar diffusion-weighted imaging (SS-EPI DWI) in cervical cancer. MATERIALS AND METHODS The RESOLVE and SS-EPI DWI images of 36 patients with histopathologically confirmed cervical cancer were collected retrospectively. Two observers independently delineated the whole tumour on RESOLVE and SS-EPI DWI, and then copied them to the corresponding ADC maps. Shape, first-order, and texture features were extracted from ADC maps in the original and filtered (Laplacian of Gaussian [LoG] and wavelet) images. Thereafter, 1,316 features were generated in each RESOLVE and SS-EPI DWI, respectively. The reproducibility of radiomic features was assessed using intraclass correlation coefficient (ICC). RESULTS In the original images, RESOLVE showed 92.86%, 66.67%, and 86.67% of features with excellent reproducibility in shape, first-order, and texture features, while SS-EPI DWI showed 85.71%, 72.22%, and 60% of features with excellent reproducibility, respectively. In the LoG and wavelet filtered images, RESOLVE had 56.77% and 65.32% of features with excellent reproducibility and SS-EPI DWI had 44.95% and 61.96% of features with excellent reproducibility, respectively. CONCLUSION Compared with SS-EPI DWI, the feature reproducibility of RESOLVE was better in cervical cancer, especially for texture features. The filtered images cannot improve the feature reproducibility compared with the original images for both SS-EPI DWI and RESOLVE.
Collapse
|
28
|
Cárcamo Ibarra PM, López González UA, Esteban Hurtado A, Navas de la Cruz MA, Asensio Valero L, Diez Domingo S. Progress and current utility of radiomics in PET/CT study of non-metastatic breast cancer: A systematic review. Rev Esp Med Nucl Imagen Mol 2023; 42:83-92. [PMID: 36375751 DOI: 10.1016/j.remnie.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022]
Abstract
AIM To synthesize the current evidence of the usefulness of radiomics in PET/CT image analysis in local and locally advanced breast cancer. Also, to evaluate the methodological quality of the radiomic studies published. METHODS Systematic review of articles in different databases until 2021 using the terms "PET", "radiomics", "texture", "breast". Only articles with human data and that included a PET image were included. Studies with simulated data and with less than 20 patients were excluded. Were extracted sample size, radiotracer used, imaging technique, and radiomics characteristics from each article. The methodological quality of the studies was determined using the QUADAS-2 tool. RESULTS 18 articles were selected. The retrospective design was the most used. The most studied radiomic characteristic was SUVmax. Several radiomic parameters were correlated with tumor characterization, and tumor heterogeneity proved useful for predicting disease course and response to treatment. Most articles showed a high risk of bias, mainly from the patient selection. CONCLUSIONS A high probability of bias was observed in most of the published articles. Radiomics is a developing field and more studies are needed to demonstrate its usefulness in routine clinical practice. The QUADAS-2 tool allows critical assessment of the methodological quality of the available evidence. Despite its limitations, radiomics is shown to be an instrument that can help to achieve personalized oncologic management of breast cancer.
Collapse
Affiliation(s)
- P M Cárcamo Ibarra
- Servicio de Medicina Nuclear, Hospital Clínico Universitario de Valencia, Spain
| | - U A López González
- Servicio de Medicina Preventiva, Hospital Universitario Doctor Peset, Valencia, Spain
| | - A Esteban Hurtado
- Servicio de Medicina Nuclear, Hospital Universitario Doctor Peset, Valencia, Spain
| | - M A Navas de la Cruz
- Servicio de Medicina Nuclear, Hospital Universitario Doctor Peset, Valencia, Spain
| | - L Asensio Valero
- Servicio de Medicina Nuclear, Hospital Clínico Universitario de Valencia, Spain
| | - S Diez Domingo
- Servicio de Protección Radiológica, Hospital Clínico Universitario de Valencia, Valencia, Spain.
| |
Collapse
|
29
|
Mazzara S, Travaini L, Botta F, Granata C, Motta G, Melle F, Fiori S, Tabanelli V, Vanazzi A, Ramadan S, Radice T, Raimondi S, Lo Presti G, Ferrari ME, Jereczek-Fossa BA, Tarella C, Ceci F, Pileri S, Derenzini E. Gene expression profiling and FDG-PET radiomics uncover radiometabolic signatures associated with outcome in DLBCL. Blood Adv 2023; 7:630-643. [PMID: 36806558 PMCID: PMC9979764 DOI: 10.1182/bloodadvances.2022007825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/06/2022] [Indexed: 02/23/2023] Open
Abstract
Emerging evidence indicates that chemoresistance is closely related to altered metabolism in cancer. Here, we hypothesized that distinct metabolic gene expression profiling (GEP) signatures might be correlated with outcome and with specific fluorodeoxyglucose positron emission tomography (FDG-PET) radiomic profiles in diffuse large B-cell lymphoma (DLBCL). We retrospectively analyzed a discovery cohort of 48 consecutive patients with DLBCL treated at our center with standard first-line chemoimmunotherapy by performing targeted GEP (T-GEP)- and FDG-PET radiomic analyses on the same target lesions at baseline. T-GEP-based metabolic profiling identified a 6-gene signature independently associated with outcomes in univariate and multivariate analyses. This signature included genes regulating mitochondrial oxidative metabolism (SCL25A1, PDK4, PDPR) that were upregulated and was inversely associated with genes involved in hypoxia and glycolysis (MAP2K1, HIF1A, GBE1) that were downregulated. These data were validated in 2 large publicly available cohorts. By integrating FDG-PET radiomics and T-GEP, we identified a radiometabolic signature (RadSig) including 4 radiomic features (histo kurtosis, histo energy, shape sphericity, and neighboring gray level dependence matrix contrast), significantly associated with the metabolic GEP-based signature (r = 0.43, P = .0027) and with progression-free survival (P = .028). These results were confirmed using different target lesions, an alternative segmentation method, and were validated in an independent cohort of 64 patients. RadSig retained independent prognostic value in relation to the International Prognostic Index score and metabolic tumor volume (MTV). Integration of RadSig and MTV further refined prognostic stratification. This study provides the proof of principle for the use of FDG-PET radiomics as a tool for noninvasive assessment of cancer metabolism and prognostic stratification in DLBCL.
Collapse
Affiliation(s)
- Saveria Mazzara
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | | | | | | - Giovanna Motta
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Federica Melle
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Stefano Fiori
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Valentina Tabanelli
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Anna Vanazzi
- Oncohematology Division, IEO IRCCS, Milan, Italy
| | - Safaa Ramadan
- Oncohematology Division, IEO IRCCS, Milan, Italy
- NCI-Cairo University, Cairo, Egypt
| | | | - Sara Raimondi
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, Milan, Italy
| | - Giuliana Lo Presti
- Molecular and Pharmaco-Epidemiology Unit, Department of Experimental Oncology, IEO IRCCS, Milan, Italy
| | | | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Francesco Ceci
- Nuclear Medicine Division, IEO IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefano Pileri
- Haematopathology Division, European Institute of Oncology (IEO) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Enrico Derenzini
- Oncohematology Division, IEO IRCCS, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
30
|
Aweda TA, Cheng SH, Lenhard SC, Sepp A, Skedzielewski T, Hsu CY, Marshall S, Haag H, Kehler J, Jagdale P, Peter A, Schmid MA, Gehman A, Doan M, Mayer AP, Gorycki P, Fanget M, Colas C, Smith B, Maier CC, Alsaid H. In vivo biodistribution and pharmacokinetics of sotrovimab, a SARS-CoV-2 monoclonal antibody, in healthy cynomolgus monkeys. Eur J Nucl Med Mol Imaging 2023; 50:667-678. [PMID: 36305907 PMCID: PMC9614201 DOI: 10.1007/s00259-022-06012-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/16/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE Sotrovimab (VIR-7831), a human IgG1κ monoclonal antibody (mAb), binds to a conserved epitope on the SARS-CoV-2 spike protein receptor binding domain (RBD). The Fc region of VIR-7831 contains an LS modification to promote neonatal Fc receptor (FcRn)-mediated recycling and extend its serum half-life. Here, we aimed to evaluate the impact of the LS modification on tissue biodistribution, by comparing VIR-7831 to its non-LS-modified equivalent, VIR-7831-WT, in cynomolgus monkeys. METHODS 89Zr-based PET/CT imaging of VIR-7831 and VIR-7831-WT was performed up to 14 days post injection. All major organs were analyzed for absolute concentration as well as tissue:blood ratios, with the focus on the respiratory tract, and a physiologically based pharmacokinetics (PBPK) model was used to evaluate the tissue biodistribution kinetics. Radiomics features were also extracted from the PET images and SUV values. RESULTS SUVmean uptake in the pulmonary bronchi for 89Zr-VIR-7831 was statistically higher than for 89Zr-VIR-7831-WT at days 6 (3.43 ± 0.55 and 2.59 ± 0.38, respectively) and 10 (2.66 ± 0.32 and 2.15 ± 0.18, respectively), while the reverse was observed in the liver at days 6 (5.14 ± 0.80 and 8.63 ± 0.89, respectively), 10 (4.52 ± 0.59 and 7.73 ± 0.66, respectively), and 14 (4.95 ± 0.65 and 7.94 ± 0.54, respectively). Though the calculated terminal half-life was 21.3 ± 3.0 days for VIR-7831 and 16.5 ± 1.1 days for VIR-7831-WT, no consistent differences were observed in the tissue:blood ratios between the antibodies except in the liver. While the lung:blood SUVmean uptake ratio for both mAbs was 0.25 on day 3, the PBPK model predicted the total lung tissue and the interstitial space to serum ratio to be 0.31 and 0.55, respectively. Radiomics analysis showed VIR-7831 had mean-centralized PET SUV distribution in the lung and liver, indicating more uniform uptake than VIR-7831-WT. CONCLUSION The half-life extended VIR-7831 remained in circulation longer than VIR-7831-WT, consistent with enhanced FcRn binding, while the tissue:blood concentration ratios in most tissues for both drugs remained statistically indistinguishable throughout the course of the experiment. In the bronchiolar region, a higher concentration of 89Zr-VIR-7831 was detected. The data also allow unparalleled insight into tissue distribution and elimination kinetics of mAbs that can guide future biologic drug discovery efforts, while the residualizing nature of the 89Zr label sheds light on the sites of antibody catabolism.
Collapse
Affiliation(s)
- Tolulope A Aweda
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Shih-Hsun Cheng
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Stephen C Lenhard
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | | | | | - Chih-Yang Hsu
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Shelly Marshall
- Integrated Biological Platform Sciences, GSK, Collegeville, PA, USA
| | - Heather Haag
- Integrated Biological Platform Sciences, GSK, Collegeville, PA, USA
| | - Jonathan Kehler
- Bioanalysis, Immunogenicity & Biomarkers, GSK, Collegeville, PA, USA
| | | | - Alessia Peter
- mAb Engineering & Bioanalytics, Humabs BioMed SA, Vir Biotechnology, Inc, Bellinzona, Switzerland
| | - Michael A Schmid
- mAb Engineering & Bioanalytics, Humabs BioMed SA, Vir Biotechnology, Inc, Bellinzona, Switzerland
| | - Andrew Gehman
- Non-Clinical and Translational Statistics, GSK, Collegeville, PA, USA
| | - Minh Doan
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA
| | - Andrew P Mayer
- Bioanalysis, Immunogenicity & Biomarkers, GSK, Collegeville, PA, USA
| | | | - Marie Fanget
- Bioanalytical Department, Vir Biotechnology, Inc, San Francisco, CA, USA
| | | | - Brenda Smith
- Toxicology, Vir Biotechnology, Inc, San Francisco, CA, USA
| | | | - Hasan Alsaid
- Bioimaging, GSK, 1250 S. Collegeville Rd, Collegeville, PA, 19426, USA.
| |
Collapse
|
31
|
Hatt M, Krizsan AK, Rahmim A, Bradshaw TJ, Costa PF, Forgacs A, Seifert R, Zwanenburg A, El Naqa I, Kinahan PE, Tixier F, Jha AK, Visvikis D. Joint EANM/SNMMI guideline on radiomics in nuclear medicine : Jointly supported by the EANM Physics Committee and the SNMMI Physics, Instrumentation and Data Sciences Council. Eur J Nucl Med Mol Imaging 2023; 50:352-375. [PMID: 36326868 PMCID: PMC9816255 DOI: 10.1007/s00259-022-06001-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE The purpose of this guideline is to provide comprehensive information on best practices for robust radiomics analyses for both hand-crafted and deep learning-based approaches. METHODS In a cooperative effort between the EANM and SNMMI, we agreed upon current best practices and recommendations for relevant aspects of radiomics analyses, including study design, quality assurance, data collection, impact of acquisition and reconstruction, detection and segmentation, feature standardization and implementation, as well as appropriate modelling schemes, model evaluation, and interpretation. We also offer an outlook for future perspectives. CONCLUSION Radiomics is a very quickly evolving field of research. The present guideline focused on established findings as well as recommendations based on the state of the art. Though this guideline recognizes both hand-crafted and deep learning-based radiomics approaches, it primarily focuses on the former as this field is more mature. This guideline will be updated once more studies and results have contributed to improved consensus regarding the application of deep learning methods for radiomics. Although methodological recommendations in the present document are valid for most medical image modalities, we focus here on nuclear medicine, and specific recommendations when necessary are made for PET/CT, PET/MR, and quantitative SPECT.
Collapse
Affiliation(s)
- M Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | | | - A Rahmim
- Departments of Radiology and Physics, University of British Columbia, Vancouver, BC, Canada
| | - T J Bradshaw
- Department of Radiology, University of Wisconsin, Madison, WI, USA
| | - P F Costa
- Department of Nuclear Medicine, West German Cancer Center, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | | | - R Seifert
- Department of Nuclear Medicine, West German Cancer Center, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany.
- Department of Nuclear Medicine, Münster University Hospital, Münster, Germany.
| | - A Zwanenburg
- 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/UCC), Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - I El Naqa
- Department of Machine Learning, Moffitt Cancer Center, Tampa, FL, 33626, USA
| | - P E Kinahan
- Imaging Research Laboratory, PET/CT Physics, Department of Radiology, UW Medical Center, University of Washington, Seattle, WA, USA
| | - F Tixier
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - A K Jha
- McKelvey School of Engineering and Mallinckrodt Institute of Radiology, Washington University in St. Louis, Saint Louis, MO, USA
| | - D Visvikis
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| |
Collapse
|
32
|
Soleymani Y, Jahanshahi AR, Pourfarshid A, Khezerloo D. Reproducibility assessment of radiomics features in various ultrasound scan settings and different scanner vendors. J Med Imaging Radiat Sci 2022; 53:664-671. [PMID: 36266173 DOI: 10.1016/j.jmir.2022.09.018] [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: 03/19/2022] [Revised: 09/09/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Radiomics in Ultrasound (US) imaging has been investigated for the prediction and prognosis of cancers. However, inter-scanner and intra-scanner variations may affect the reproducibility of radiomics results. This study aims to evaluate the reproducibility of US textural radiomics features across various scan settings and scanner vendors. MATERIALS AND METHODS US images in quality control (QC) phantom were obtained by three scanners (Philips, Samsung, and Siemens) with different scan settings and parameters. Circular regions of interest (ROIs) inside isoechoic, hypoechoic, and hyperechoic objects were manually delineated. Forty textural radiomics features were extracted from each ROI, and then the robust features that could distinguish different echogenic objects were obtained by the Mann-Whitney U test. Reproducibility of the robust radiomics features was assessed by the intraclass correlation coefficient (ICC) and coefficient of variation (CV); ICC>0.90 and %CV<20 were considered reproducible. RESULTS According to the Mann-Whitney U test results, ten robust features could differentiate the hypoechoic, and 15 robust features could differentiate the hyperechoic objects from the isoechoic objects (P<0.001). The total ICC of the robust features for each echogenic object was >0.95 in different scanners and scan settings. Four and seven features were individually reproducible (%CV < 20, ICC>0.90) in hypoechoic and hyperechoic objects, respectively. Also, four features seem reproducible by changing the ROI location across the horizontal and vertical lines for both convex and linear array transducers. CONCLUSIONS Most of the US textural radiomics features in this study were not reproducible. However, several features showed high reproducibility at different scan settings and scanners. These features may also be reproducible when ROI size and location change slightly.
Collapse
Affiliation(s)
- Yunus Soleymani
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Reza Jahanshahi
- Tabriz University of Medical Sciences, Faculty of Medicine, Imam Reza Hospital, Department of Radiology
| | - Amin Pourfarshid
- Department of Medical Physics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Davood Khezerloo
- Department of Radiology, Faculty of Alliance Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran & Medical Radiation Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
33
|
Muacevic A, Adler JR, Issa M, Ali O, Noureldin K, Gaber A, Mahgoub A, Ahmed M, Yousif M, Zeinaldine A. Textural Analysis as a Predictive Biomarker in Rectal Cancer. Cureus 2022; 14:e32241. [PMID: 36620843 PMCID: PMC9813797 DOI: 10.7759/cureus.32241] [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] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a common deadly cancer. Early detection and accurate staging of CRC enhance good prognosis and better treatment outcomes. Rectal cancer staging is the cornerstone for selecting the best treatment approach. The standard gold method for rectal cancer staging is pelvic MRI. After staging, combining surgery and chemoradiation is the standard management aiming for a curative outcome. Textural analysis (TA) is a radiomic process that quantifies lesions' heterogenicity by measuring pixel distribution in digital imaging. MRI textural analysis (MRTA) of rectal cancer images is growing in current literature as a future predictor of outcomes of rectal cancer management, such as pathological response to neoadjuvant chemoradiotherapy (NCRT), survival, and tumour recurrence. MRTA techniques could validate alternative approaches in rectal cancer treatment, such as the wait-and-watch (W&W) approach in pathologically complete responders (pCR) following NCRT. We consider this a significant step towards implementing precision management in rectal cancer. In this narrative review, we summarize the current knowledge regarding the potential role of TA in rectal cancer management in predicting the prognosis and clinical outcomes, as well as aim to delineate the challenges which obstruct the implementing of this new modality in clinical practice.
Collapse
|
34
|
Alsaid H, Cheng SH, Bi M, Xie F, Rambo M, Skedzielewski T, Hoang B, Mohanan S, Comroe D, Gehman A, Hsu CY, Farhangi K, Tran H, Sherina V, Doan M, Groseclose MR, Hopson CB, Brett S, Wilson IA, Nicholls A, Ballas M, Waight JD, Jucker BM. Immuno-PET Monitoring of CD8 + T Cell Infiltration Post ICOS Agonist Antibody Treatment Alone and in Combination with PD-1 Blocking Antibody Using a 89Zr Anti-CD8 + Mouse Minibody in EMT6 Syngeneic Tumor Mouse. Mol Imaging Biol 2022; 25:528-540. [PMID: 36266600 PMCID: PMC10172244 DOI: 10.1007/s11307-022-01781-7] [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: 05/29/2022] [Revised: 09/15/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE The presence and functional competence of intratumoral CD8+ T cells is often a barometer for successful immunotherapeutic responses in cancer. Despite this understanding and the extensive number of clinical-stage immunotherapies focused on potentiation (co-stimulation) or rescue (checkpoint blockade) of CD8+ T cell antitumor activity, dynamic biomarker strategies are often lacking. To help fill this gap, immuno-PET nuclear imaging has emerged as a powerful tool for in vivo molecular imaging of antibody targeting. Here, we took advantage of immuno-PET imaging using 89Zr-IAB42M1-14, anti-mouse CD8 minibody, to characterize CD8+ T-cell tumor infiltration dynamics following ICOS (inducible T-cell co-stimulator) agonist antibody treatment alone and in combination with PD-1 blocking antibody in a model of mammary carcinoma. PROCEDURES Female BALB/c mice with established EMT6 tumors received 10 µg, IP of either IgG control antibodies, ICOS agonist monotherapy, or ICOS/PD-1 combination therapy on days 0, 3, 5, 7, 9, 10, or 14. Imaging was performed at 24 and 48 h post IV dose of 89Zr IAB42M1-14. In addition to 89Zr-IAB42M1-14 uptake in tumor and tumor-draining lymph node (TDLN), 3D radiomic features were extracted from PET/CT images to identify treatment effects. Imaging mass cytometry (IMC) and immunohistochemistry (IHC) was performed at end of study. RESULTS 89Zr-IAB42M1-14 uptake in the tumor was observed by day 11 and was preceded by an increase in the TDLN as early as day 4. The spatial distribution of 89Zr-IAB42M1-14 was more uniform in the drug treated vs. control tumors, which had spatially distinct tracer uptake in the periphery relative to the core of the tumor. IMC analysis showed an increased percentage of cytotoxic T cells in the ICOS monotherapy and ICOS/PD-1 combination group compared to IgG controls. Additionally, temporal radiomics analysis demonstrated early predictiveness of imaging features. CONCLUSION To our knowledge, this is the first detailed description of the use of a novel immune-PET imaging technique to assess the kinetics of CD8+ T-cell infiltration into tumor and lymphoid tissues following ICOS agonist and PD-1 blocking antibody therapy. By demonstrating the capacity for increased spatial and temporal resolution of CD8+ T-cell infiltration across tumors and lymphoid tissues, these observations underscore the widespread potential clinical utility of non-invasive PET imaging for T-cell-based immunotherapy in cancer.
Collapse
Affiliation(s)
- Hasan Alsaid
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA.
| | - Shih-Hsun Cheng
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Meixia Bi
- Immuno-Oncology Research Unit, GlaxoSmithKline, Collegeville, PA, USA
| | - Fang Xie
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Mary Rambo
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | | | - Bao Hoang
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Sunish Mohanan
- Non-Clinical Safety, IVIVT, GlaxoSmithKline, Collegeville, PA, USA
| | - Debra Comroe
- Integrated Biological Platform Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Andrew Gehman
- Research Statistics, GlaxoSmithKline, Collegeville, PA, USA
| | - Chih-Yang Hsu
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Kamyar Farhangi
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | - Hoang Tran
- Research Statistics, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Minh Doan
- Bioimaging, IVIVT, GlaxoSmithKline, Collegeville, PA, 19426, USA
| | | | | | - Sara Brett
- Oncology Cell Therapy Research Unit, GlaxoSmithKline, Hertfordshire, UK
| | | | | | - Marc Ballas
- Oncology Clinical Development, GlaxoSmithKline, Collegeville, PA, USA
| | - Jeremy D Waight
- Immuno-Oncology Research Unit, GlaxoSmithKline, Collegeville, PA, USA
| | - Beat M Jucker
- Clinical Imaging, GlaxoSmithKline, Collegeville, PA, USA
| |
Collapse
|
35
|
Cárcamo Ibarra P, López González U, Esteban Hurtado A, Navas de la Cruz M, Asensio Valero L, Diez Domingo S. Progreso y utilidad actual de la radiómica dentro del estudio PET/TC en cáncer de mama no metastásico: una revisión sistemática. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
36
|
Kihira S, Mei X, Mahmoudi K, Liu Z, Dogra S, Belani P, Tsankova N, Hormigo A, Fayad ZA, Doshi A, Nael K. U-Net Based Segmentation and Characterization of Gliomas. Cancers (Basel) 2022; 14:4457. [PMID: 36139616 PMCID: PMC9496685 DOI: 10.3390/cancers14184457] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/18/2022] Open
Abstract
(1) Background: Gliomas are the most common primary brain neoplasms accounting for roughly 40−50% of all malignant primary central nervous system tumors. We aim to develop a deep learning-based framework for automated segmentation and prediction of biomarkers and prognosis in patients with gliomas. (2) Methods: In this retrospective two center study, patients were included if they (1) had a diagnosis of glioma with known surgical histopathology and (2) had preoperative MRI with FLAIR sequence. The entire tumor volume including FLAIR hyperintense infiltrative component and necrotic and cystic components was segmented. Deep learning-based U-Net framework was developed based on symmetric architecture from the 512 × 512 segmented maps from FLAIR as the ground truth mask. (3) Results: The final cohort consisted of 208 patients with mean ± standard deviation of age (years) of 56 ± 15 with M/F of 130/78. DSC of the generated mask was 0.93. Prediction for IDH-1 and MGMT status had a performance of AUC 0.88 and 0.62, respectively. Survival prediction of <18 months demonstrated AUC of 0.75. (4) Conclusions: Our deep learning-based framework can detect and segment gliomas with excellent performance for the prediction of IDH-1 biomarker status and survival.
Collapse
Affiliation(s)
- Shingo Kihira
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| | - Xueyan Mei
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Keon Mahmoudi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| | - Zelong Liu
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Siddhant Dogra
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Puneet Belani
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nadejda Tsankova
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adilia Hormigo
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zahi A. Fayad
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amish Doshi
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kambiz Nael
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90033, USA
| |
Collapse
|
37
|
Yusufaly TI, Zou J, Nelson TJ, Williamson CW, Simon A, Singhal M, Liu H, Wong H, Saenz CC, Mayadev J, McHale MT, Yashar CM, Eskander R, Sharabi A, Hoh CK, Obrzut S, Mell LK. Improved Prognosis of Treatment Failure in Cervical Cancer with Nontumor PET/CT Radiomics. J Nucl Med 2022; 63:1087-1093. [PMID: 34711618 PMCID: PMC9258568 DOI: 10.2967/jnumed.121.262618] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Radiomics has been applied to predict recurrence in several disease sites, but current approaches are typically restricted to analyzing tumor features, neglecting nontumor information in the rest of the body. The purpose of this work was to develop and validate a model incorporating nontumor radiomics, including whole-body features, to predict treatment outcomes in patients with previously untreated locoregionally advanced cervical cancer. Methods: We analyzed 127 cervical cancer patients treated definitively with chemoradiotherapy and intracavitary brachytherapy. All patients underwent pretreatment whole-body 18F-FDG PET/CT. To quantify effects due to the tumor itself, the gross tumor volume (GTV) was directly contoured on the PET/CT image. Meanwhile, to quantify effects arising from the rest of the body, the planning target volume (PTV) was deformably registered from each planning CT to the PET/CT scan, and a semiautomated approach combining seed-growing and manual contour review generated whole-body muscle, bone, and fat segmentations on each PET/CT image. A total of 965 radiomic features were extracted for GTV, PTV, muscle, bone, and fat. Ninety-five patients were used to train a Cox model of disease recurrence including both radiomic and clinical features (age, stage, tumor grade, histology, and baseline complete blood cell counts), using bagging and split-sample-validation for feature reduction and model selection. To further avoid overfitting, the resulting models were tested for generalization on the remaining 32 patients, by calculating a risk score based on Cox regression and evaluating the c-index (c-index > 0.5 indicates predictive power). Results: Optimal performance was seen in a Cox model including 1 clinical biomarker (whether or not a tumor was stage III-IVA), 2 GTV radiomic biomarkers (PET gray-level size-zone matrix small area low gray level emphasis and zone entropy), 1 PTV radiomic biomarker (major axis length), and 1 whole-body radiomic biomarker (CT bone root mean square). In particular, stratification into high- and low-risk groups, based on the linear risk score from this Cox model, resulted in a hazard ratio of 0.019 (95% CI, 0.004, 0.082), an improvement over stratification based on clinical stage alone, which had a hazard ratio of 0.36 (95% CI, 0.16, 0.83). Conclusion: Incorporating nontumor radiomic biomarkers can improve the performance of prognostic models compared with using only clinical and tumor radiomic biomarkers. Future work should look to further test these models in larger, multiinstitutional cohorts.
Collapse
Affiliation(s)
- Tahir I Yusufaly
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins University, School of Medicine, Baltimore, Maryland;
| | - Jingjing Zou
- Department of Family Medicine and Public Health and Department of Mathematics, University of California San Diego, La Jolla, California
| | - Tyler J Nelson
- Center for Precision Radiation Medicine, La Jolla, California
| | - Casey W Williamson
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Aaron Simon
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | | | - Hannah Liu
- Center for Precision Radiation Medicine, La Jolla, California
| | - Hank Wong
- Center for Precision Radiation Medicine, La Jolla, California
| | - Cheryl C Saenz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, University of California San Diego, La Jolla, California; and
| | - Jyoti Mayadev
- Center for Precision Radiation Medicine, La Jolla, California
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Michael T McHale
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, University of California San Diego, La Jolla, California; and
| | - Catheryn M Yashar
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Ramez Eskander
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Gynecologic Oncology, University of California San Diego, La Jolla, California; and
| | - Andrew Sharabi
- Center for Precision Radiation Medicine, La Jolla, California
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| | - Carl K Hoh
- Department of Radiology, Division of Nuclear Medicine, University of California San Diego, La Jolla, California
| | - Sebastian Obrzut
- Department of Radiology, Division of Nuclear Medicine, University of California San Diego, La Jolla, California
| | - Loren K Mell
- Center for Precision Radiation Medicine, La Jolla, California
- Department of Radiation Medicine and Applied Sciences, University of California San Diego, La Jolla, California
| |
Collapse
|
38
|
Karahan Şen NP, Alataş Ö, Gülcü A, Özdoğan Ö, Derebek E, Çapa Kaya G. The role of volumetric and textural analysis of pretreatment 18F-fluorodeoxyglucose PET/computerized tomography images in predicting complete response to transarterial radioembolization in hepatocellular cancer. Nucl Med Commun 2022; 43:807-814. [PMID: 35506284 DOI: 10.1097/mnm.0000000000001572] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study evaluates the role of pretreatment 18F-FDG PET/CT in predicting the response to treatment in patients with hepatocellular cancer (HCC) who applied transarterial radioembolization (TARE) via the volumetric and texture features extracted from 18F-FDG PET/CT images. METHODS Thirty-three patients with HCC who had applied TARE [lobar (LT) or superselective (ST)] after 18F-FDG PET/CT were included in the study. Response to the treatment was evaluated from posttherapy magnetic resonance (MR). Patients were divided into two groups: the responder group (RG) (complete responders) and non-RG (NRG) (including partial response, stabile, and progressive). Metabolic tumor volume (MTV) and total lesion glycolysis (TLG) and texture features were extracted from PET/CT images. The differences among MTV, TLG, and texture features between response groups were analyzed with the Mann-Whitney U test. ROC analysis was performed for features with P < 0.05. Spearman correlation analysis was used, and features with correlation coefficient < 0.8 were evaluated with the logistic regression analysis. RESULTS Significant differences were detected in TLG, MTV, SHAPE_compacity, GLCM_correlation, GLRLM_GLNU, GLRLM_RLNU, NGLDM_coarseness, NGLDM_busyness, GLZLM_LZHGE, GLZLM_GLNU, and GLZLM_ZLNU between RG and NRG. Multivariate analysis demonstrated that MTV was the only meaningful parameter with an AUC of 0.827 (P = 0.002; 95% CI, 0.688-0.966). The best cutoff value was determined as 74.11 ml with 78.9% sensitivity and 78.6% specificity in discriminating nonresponders. CONCLUSION In predicting the curative effect of TARE, multivariate analysis results demonstrated that MTV was the only independent predictor, and MTV higher than 74.11 ml were determined the best predictor of nonresponders.
Collapse
Affiliation(s)
| | - Özkan Alataş
- Radiology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aytaç Gülcü
- Radiology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | | | | | | |
Collapse
|
39
|
Zschaeck S, Weingärtner J, Lombardo E, Marschner S, Hajiyianni M, Beck M, Zips D, Li Y, Lin Q, Amthauer H, Troost EGC, van den Hoff J, Budach V, Kotzerke J, Ferentinos K, Karagiannis E, Kaul D, Gregoire V, Holzgreve A, Albert NL, Nikulin P, Bachmann M, Kopka K, Krause M, Baumann M, Kazmierska J, Cegla P, Cholewinski W, Strouthos I, Zöphel K, Majchrzak E, Landry G, Belka C, Stromberger C, Hofheinz F. 18F-Fluorodeoxyglucose Positron Emission Tomography of Head and Neck Cancer: Location and HPV Specific Parameters for Potential Treatment Individualization. Front Oncol 2022; 12:870319. [PMID: 35756665 PMCID: PMC9213669 DOI: 10.3389/fonc.2022.870319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022] Open
Abstract
Purpose 18F-fluorodeoxyglucose positron emission tomography (FDG-PET) is utilized for staging and treatment planning of head and neck squamous cell carcinomas (HNSCC). Some older publications on the prognostic relevance showed inconclusive results, most probably due to small study sizes. This study evaluates the prognostic and potentially predictive value of FDG-PET in a large multi-center analysis. Methods Original analysis of individual FDG-PET and patient data from 16 international centers (8 institutional datasets, 8 public repositories) with 1104 patients. All patients received curative intent radiotherapy/chemoradiation (CRT) and pre-treatment FDG-PET imaging. Primary tumors were semi-automatically delineated for calculation of SUVmax, SUVmean, metabolic tumor volume (MTV) and total lesion glycolysis (TLG). Cox regression analyses were performed for event-free survival (EFS), overall survival (OS), loco-regional control (LRC) and freedom from distant metastases (FFDM). Results FDG-PET parameters were associated with patient outcome in the whole cohort regarding clinical endpoints (EFS, OS, LRC, FFDM), in uni- and multivariate Cox regression analyses. Several previously published cut-off values were successfully validated. Subgroup analyses identified tumor- and human papillomavirus (HPV) specific parameters. In HPV positive oropharynx cancer (OPC) SUVmax was well suited to identify patients with excellent LRC for organ preservation. Patients with SUVmax of 14 or less were unlikely to develop loco-regional recurrence after definitive CRT. In contrast FDG PET parameters deliver only limited prognostic information in laryngeal cancer. Conclusion FDG-PET parameters bear considerable prognostic value in HNSCC and potential predictive value in subgroups of patients, especially regarding treatment de-intensification and organ-preservation. The potential predictive value needs further validation in appropriate control groups. Further research on advanced imaging approaches including radiomics or artificial intelligence methods should implement the identified cut-off values as benchmark routine imaging parameters.
Collapse
Affiliation(s)
- Sebastian Zschaeck
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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
| | - Julian Weingärtner
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Elia Lombardo
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Sebastian Marschner
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Marina Hajiyianni
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcus Beck
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniel Zips
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany, Germany.,Department of Radiation Oncology, University Hospital and Medical Faculty, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Yimin Li
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qin Lin
- Department of Radiation Oncology, Xiamen Cancer Center, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Holger Amthauer
- Department of Nuclear Medicine, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, 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.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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.,Institute of Radiooncology - OncoRay, 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
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Volker Budach
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jörg Kotzerke
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden, Germany
| | - Konstantinos Ferentinos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Efstratios Karagiannis
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - David Kaul
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Vincent Gregoire
- Radiation Oncology Department, Leon Bérard Cancer Center, Lyon, France
| | - Adrien Holzgreve
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Germany
| | - Pavel Nikulin
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Mechthild Krause
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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.,Institute of Radiooncology - OncoRay, 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
| | - Michael Baumann
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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.,Institute of Radiooncology - OncoRay, Helmholtz-Zentrum Dresden - Rossendorf, Dresden, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joanna Kazmierska
- Electroradiology Department, University of Medical Sciences, Poznan, Poland.,Radiotherapy Department II, Greater Poland Cancer Centre, Poznan, Poland
| | - Paulina Cegla
- Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Witold Cholewinski
- Electroradiology Department, University of Medical Sciences, Poznan, Poland.,Department of Nuclear Medicine, Greater Poland Cancer Centre, Poznan, Poland
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Klaus Zöphel
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ) Heidelberg, Germany, 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.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Dresden, Germany.,Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Ewa Majchrzak
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Greater Poland Cancer Centre, Poznan, Poland
| | - Guillaume Landry
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, Ludwig-Maximilians-University (LMU) Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Carmen Stromberger
- Department of Radiation Oncology, Berlin Institute of Health, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank Hofheinz
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| |
Collapse
|
40
|
Abdollahi H, Chin E, Clark H, Hyde DE, Thomas S, Wu J, Uribe CF, Rahmim A. Radiomics-guided radiation therapy: opportunities and challenges. Phys Med Biol 2022; 67. [DOI: 10.1088/1361-6560/ac6fab] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/13/2022] [Indexed: 11/11/2022]
Abstract
Abstract
Radiomics is an advanced image-processing framework, which extracts image features and considers them as biomarkers towards personalized medicine. Applications include disease detection, diagnosis, prognosis, and therapy response assessment/prediction. As radiation therapy aims for further individualized treatments, radiomics could play a critical role in various steps before, during and after treatment. Elucidation of the concept of radiomics-guided radiation therapy (RGRT) is the aim of this review, attempting to highlight opportunities and challenges underlying the use of radiomics to guide clinicians and physicists towards more effective radiation treatments. This work identifies the value of RGRT in various steps of radiotherapy from patient selection to follow-up, and subsequently provides recommendations to improve future radiotherapy using quantitative imaging features.
Collapse
|
41
|
Liberini V, Laudicella R, Balma M, Nicolotti DG, Buschiazzo A, Grimaldi S, Lorenzon L, Bianchi A, Peano S, Bartolotta TV, Farsad M, Baldari S, Burger IA, Huellner MW, Papaleo A, Deandreis D. Radiomics and artificial intelligence in prostate cancer: new tools for molecular hybrid imaging and theragnostics. Eur Radiol Exp 2022; 6:27. [PMID: 35701671 PMCID: PMC9198151 DOI: 10.1186/s41747-022-00282-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022] Open
Abstract
In prostate cancer (PCa), the use of new radiopharmaceuticals has improved the accuracy of diagnosis and staging, refined surveillance strategies, and introduced specific and personalized radioreceptor therapies. Nuclear medicine, therefore, holds great promise for improving the quality of life of PCa patients, through managing and processing a vast amount of molecular imaging data and beyond, using a multi-omics approach and improving patients’ risk-stratification for tailored medicine. Artificial intelligence (AI) and radiomics may allow clinicians to improve the overall efficiency and accuracy of using these “big data” in both the diagnostic and theragnostic field: from technical aspects (such as semi-automatization of tumor segmentation, image reconstruction, and interpretation) to clinical outcomes, improving a deeper understanding of the molecular environment of PCa, refining personalized treatment strategies, and increasing the ability to predict the outcome. This systematic review aims to describe the current literature on AI and radiomics applied to molecular imaging of prostate cancer.
Collapse
Affiliation(s)
- Virginia Liberini
- Medical Physiopathology - A.O.U. Città della Salute e della Scienza di Torino, Division of Nuclear Medicine, Department of Medical Science, University of Torino, 10126, Torino, Italy. .,Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy.
| | - Riccardo Laudicella
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006, Zurich, Switzerland.,Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, University of Messina, 98125, Messina, Italy.,Nuclear Medicine Unit, Fondazione Istituto G. Giglio, Ct.da Pietrapollastra Pisciotto, Cefalù, Palermo, Italy
| | - Michele Balma
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy
| | | | - Ambra Buschiazzo
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy
| | - Serena Grimaldi
- Medical Physiopathology - A.O.U. Città della Salute e della Scienza di Torino, Division of Nuclear Medicine, Department of Medical Science, University of Torino, 10126, Torino, Italy
| | - Leda Lorenzon
- Medical Physics Department, Central Bolzano Hospital, 39100, Bolzano, Italy
| | - Andrea Bianchi
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy
| | - Simona Peano
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy
| | | | - Mohsen Farsad
- Nuclear Medicine, Central Hospital Bolzano, 39100, Bolzano, Italy
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, University of Messina, 98125, Messina, Italy
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006, Zurich, Switzerland.,Department of Nuclear Medicine, Kantonsspital Baden, 5004, Baden, Switzerland
| | - Martin W Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006, Zurich, Switzerland
| | - Alberto Papaleo
- Nuclear Medicine Department, S. Croce e Carle Hospital, 12100, Cuneo, Italy
| | - Désirée Deandreis
- Medical Physiopathology - A.O.U. Città della Salute e della Scienza di Torino, Division of Nuclear Medicine, Department of Medical Science, University of Torino, 10126, Torino, Italy
| |
Collapse
|
42
|
Lumbroso J. Functional imaging for evaluation of cancers and biologically conformal radiotherapy: Past-history and present-day perspectives. Presse Med 2022; 51:104124. [PMID: 35500752 DOI: 10.1016/j.lpm.2022.104124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/17/2022] [Accepted: 04/11/2022] [Indexed: 12/16/2022] Open
Abstract
Over the past twenty years, nuclear medicine has enhanced the role of functional imaging in cancerology. A major milestone was achieved in the early 2000s with widespread availability of the positron emitter tracer 18F- deoxyglucose (FDG) and the introduction of hybrid imagers, i.e. positron imagers coupled with an X CT, providing anatomical landmarks and potently contributing to attenuation and scatter correction of the images. Other technical advances have progressively increased the quality of positron images. To date, the most widely used tracer remains FDG, which is highly beneficial in terms of sensitivity and specificity in detection of tumor sites, also providing biological information on tumors and early evaluation of treatment response for most cancers. Other highly specific tracers have been developed and are now routinely used for pheochromocytoma and paraganglioma, neuroendocrine tumors, and prostate cancer. Biological Radiotherapy has two aspects: Internal radiotherapy consisting in administration of a tumor-specific molecule radiolabeled with an isotope delivering an adequate radiation dose to the targeted tumor sites (on the model of thyroid cancer treated with radioiodine) and external radiotherapy designed to determine tumor volume, assess response and to dose radiation according to the tumor characteristics shown by functional imaging.
Collapse
Affiliation(s)
- Jean Lumbroso
- Service de Médecine Nucléaire, Gustave Roussy Cancer Campus, 114, rue Édouard-Vaillant, Villejuif 94805 CEDEX, France.
| |
Collapse
|
43
|
Chang C, Ruan M, Lei B, Yu H, Zhao W, Ge Y, Duan S, Teng W, Wu Q, Qian X, Wang L, Yan H, Liu C, Liu L, Feng J, Xie W. Development of a PET/CT molecular radiomics-clinical model to predict thoracic lymph node metastasis of invasive lung adenocarcinoma ≤ 3 cm in diameter. EJNMMI Res 2022; 12:23. [PMID: 35445899 PMCID: PMC9023644 DOI: 10.1186/s13550-022-00895-x] [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: 11/28/2021] [Accepted: 04/05/2022] [Indexed: 12/25/2022] Open
Abstract
Background To investigate the value of 18F-FDG PET/CT molecular radiomics combined with a clinical model in predicting thoracic lymph node metastasis (LNM) in invasive lung adenocarcinoma (≤ 3 cm). Methods A total of 528 lung adenocarcinoma patients were enrolled in this retrospective study. Five models were developed for the prediction of thoracic LNM, including PET radiomics, CT radiomics, PET/CT radiomics, clinical and integrated PET/CT radiomics-clinical models. Ten PET/CT radiomics features and two clinical characteristics were selected for the construction of the integrated PET/CT radiomics-clinical model. The predictive performance of all models was examined by receiver operating characteristic (ROC) curve analysis, and clinical utility was validated by nomogram analysis and decision curve analysis (DCA). Results According to ROC curve analysis, the integrated PET/CT molecular radiomics-clinical model outperformed the clinical model and the three other radiomics models, and the area under the curve (AUC) values of the integrated model were 0.95 (95% CI: 0.93–0.97) in the training group and 0.94 (95% CI: 0.89–0.97) in the test group. The nomogram analysis and DCA confirmed the clinical application value of this integrated model in predicting thoracic LNM. Conclusions The integrated PET/CT molecular radiomics-clinical model proposed in this study can ensure a higher level of accuracy in predicting the thoracic LNM of clinical invasive lung adenocarcinoma (≤ 3 cm) compared with the radiomics model or clinical model alone. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00895-x.
Collapse
Affiliation(s)
- Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Maomei Ruan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Bei Lei
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenlu Zhao
- Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yaqiong Ge
- GE Healthcare China, Pudong New Town, Shanghai, China
| | - Shaofeng Duan
- GE Healthcare China, Pudong New Town, Shanghai, China
| | - Wenjing Teng
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qianfu Wu
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaohua Qian
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lihua Wang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Hui Yan
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Ciyi Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China
| | - Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China.,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Jian Feng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, No. 241 West Huaihai Road, Shanghai, 200030, China. .,Clinical and Translational Center in Shanghai Chest Hospital, Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, China.
| |
Collapse
|
44
|
Radiomic features as biomarkers of soft tissue paediatric sarcomas: preliminary results of a PET/MR study. Radiol Oncol 2022; 56:138-141. [PMID: 35344641 PMCID: PMC9122292 DOI: 10.2478/raon-2022-0013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/04/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Pediatric soft tissue sarcomas are rare tumors with rhabdomyosarcoma being the most frequent histotype. Diagnostic imaging plays a significant role in the evaluation of this type of tumors. Thus, aim of this study was to assess the prognostic and diagnostic value of radiomic features extracted from axial T2w images of the primary lesion in children with soft tissue sarcomas examined by PET/MR for staging. METHODS Using an open source software, each lesion was segmented and 33 radiomic features then extracted. Factor and logistic regression analyses were applied to select highly correlating features and evaluate their prognostic role, respectively. Differences in radiomic, demographics, metabolic, and laboratory variables according to tumor grade and histotype were investigated by the Students' and Chi-square tests. In case of differences the diagnostic value of the variable/s was assessed by receiver operating curves. RESULTS Eighteen children (11 female; mean age 7.8 ± 4.6-year-old) matched the inclusion criteria. The factor analysis allowed the selection of five highly correlating features which, according to regression analysis, did not influence the outcome (p > 0.05, each). The feature lmc1 was significantly higher in low grade lesions (p = 0.045) and showed 70.4% accuracy in classifying high grade tumors while the feature variance was significantly lower in rhabdomyosarcomas (p = 0.008) and showed 83.3% accuracy for this histotype. CONCLUSIONS In conclusion, our preliminary results suggest that specific radiomic features may act as biomarkers of pediatric soft tissue sarcoma grade and histotype.
Collapse
|
45
|
Anagnostopoulos AK, Gaitanis A, Gkiozos I, Athanasiadis EI, Chatziioannou SN, Syrigos KN, Thanos D, Chatziioannou AN, Papanikolaou N. Radiomics/Radiogenomics in Lung Cancer: Basic Principles and Initial Clinical Results. Cancers (Basel) 2022; 14:cancers14071657. [PMID: 35406429 PMCID: PMC8997041 DOI: 10.3390/cancers14071657] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiogenomics is a promising new approach in cancer assessment, providing an evaluation of the molecular basis of imaging phenotypes after establishing associations between radiological features and molecular features at the genomic–transcriptomic–proteomic level. This review focuses on describing key aspects of radiogenomics while discussing limitations of translatability to the clinic and possible solutions to these challenges, providing the clinician with an up-to-date handbook on how to use this new tool. Abstract Lung cancer is the leading cause of cancer-related deaths worldwide, and elucidation of its complicated pathobiology has been traditionally targeted by studies incorporating genomic as well other high-throughput approaches. Recently, a collection of methods used for cancer imaging, supplemented by quantitative aspects leading towards imaging biomarker assessment termed “radiomics”, has introduced a novel dimension in cancer research. Integration of genomics and radiomics approaches, where identifying the biological basis of imaging phenotypes is feasible due to the establishment of associations between molecular features at the genomic–transcriptomic–proteomic level and radiological features, has recently emerged termed radiogenomics. This review article aims to briefly describe the main aspects of radiogenomics, while discussing its basic limitations related to lung cancer clinical applications for clinicians, researchers and patients.
Collapse
Affiliation(s)
- Athanasios K. Anagnostopoulos
- Division of Biotechnology, Center of Systems Biology, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11525 Athens, Greece
- Correspondence:
| | - Anastasios Gaitanis
- Clinical and Translational Research, Center of Experimental Surgery, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Ioannis Gkiozos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Emmanouil I. Athanasiadis
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Sofia N. Chatziioannou
- Nuclear Medicine Division, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece;
| | - Konstantinos N. Syrigos
- Third Department of Internal Medicine, “Sotiria” Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (I.G.); (K.N.S.)
| | - Dimitris Thanos
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece; (E.I.A.); (D.T.)
| | - Achilles N. Chatziioannou
- First Department of Radiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papanikolaou
- Computational Clinical Imaging Group, Centre for the Unknown, Champalimaud Foundation, 1400-038 Lisbon, Portugal;
- Machine Learning Group, The Royal Marsden, London SM2 5MG, UK
- The Institute of Cancer Research, London SM2 5MG, UK
- Karolinska Institutet, 14186 Stockholm, Sweden
- Institute of Computer Science, FORTH, 70013 Heraklion, Greece
| |
Collapse
|
46
|
Artificial Intelligence Applications on Restaging [18F]FDG PET/CT in Metastatic Colorectal Cancer: A Preliminary Report of Morpho-Functional Radiomics Classification for Prediction of Disease Outcome. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12062941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aim of this study was to investigate the application of [18F]FDG PET/CT images-based textural features analysis to propose radiomics models able to early predict disease progression (PD) and survival outcome in metastatic colorectal cancer (MCC) patients after first adjuvant therapy. For this purpose, 52 MCC patients who underwent [18F]FDGPET/CT during the disease restaging process after the first adjuvant therapy were analyzed. Follow-up data were recorded for a minimum of 12 months after PET/CT. Radiomics features from each avid lesion in PET and low-dose CT images were extracted. A hybrid descriptive-inferential method and the discriminant analysis (DA) were used for feature selection and for predictive model implementation, respectively. The performance of the features in predicting PD was performed for per-lesion analysis, per-patient analysis, and liver lesions analysis. All lesions were again considered to assess the diagnostic performance of the features in discriminating liver lesions. In predicting PD in the whole group of patients, on PET features radiomics analysis, among per-lesion analysis, only the GLZLM_GLNU feature was selected, while three features were selected from PET/CT images data set. The same features resulted more accurately by associating CT features with PET features (AUROC 65.22%). In per-patient analysis, three features for stand-alone PET images and one feature (i.e., HUKurtosis) for the PET/CT data set were selected. Focusing on liver metastasis, in per-lesion analysis, the same analysis recognized one PET feature (GLZLM_GLNU) from PET images and three features from PET/CT data set. Similarly, in liver lesions per-patient analysis, we found three PET features and a PET/CT feature (HUKurtosis). In discrimination of liver metastasis from the rest of the other lesions, optimal results of stand-alone PET imaging were found for one feature (SUVbwmin; AUROC 88.91%) and two features for merged PET/CT features analysis (AUROC 95.33%). In conclusion, our machine learning model on restaging [18F]FDGPET/CT was demonstrated to be feasible and potentially useful in the predictive evaluation of disease progression in MCC.
Collapse
|
47
|
Anan N, Zainon R, Tamal M. A review on advances in 18F-FDG PET/CT radiomics standardisation and application in lung disease management. Insights Imaging 2022; 13:22. [PMID: 35124733 PMCID: PMC8817778 DOI: 10.1186/s13244-021-01153-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
Radiomics analysis quantifies the interpolation of multiple and invisible molecular features present in diagnostic and therapeutic images. Implementation of 18-fluorine-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) radiomics captures various disorders in non-invasive and high-throughput manner. 18F-FDG PET/CT accurately identifies the metabolic and anatomical changes during cancer progression. Therefore, the application of 18F-FDG PET/CT in the field of oncology is well established. Clinical application of 18F-FDG PET/CT radiomics in lung infection and inflammation is also an emerging field. Combination of bioinformatics approaches or textual analysis allows radiomics to extract additional information to predict cell biology at the micro-level. However, radiomics texture analysis is affected by several factors associated with image acquisition and processing. At present, researchers are working on mitigating these interrupters and developing standardised workflow for texture biomarker establishment. This review article focuses on the application of 18F-FDG PET/CT in detecting lung diseases specifically on cancer, infection and inflammation. An overview of different approaches and challenges encountered on standardisation of 18F-FDG PET/CT technique has also been highlighted. The review article provides insights about radiomics standardisation and application of 18F-FDG PET/CT in lung disease management.
Collapse
|
48
|
Nakajo M, Jinguji M, Tani A, Yano E, Hoo CK, Hirahara D, Togami S, Kobayashi H, Yoshiura T. Machine learning based evaluation of clinical and pretreatment 18F-FDG-PET/CT radiomic features to predict prognosis of cervical cancer patients. Abdom Radiol (NY) 2022; 47:838-847. [PMID: 34821963 DOI: 10.1007/s00261-021-03350-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 05/25/2021] [Accepted: 11/09/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE To examine the usefulness of machine learning to predict prognosis in cervical cancer using clinical and radiomic features of 2-deoxy-2-[18F]fluoro-D-glucose (18F-FDG) positron emission tomography/computed tomography (CT) (18F-FDG-PET/CT). METHODS This retrospective study included 50 cervical cancer patients who underwent 18F-FDG-PET/CT before treatment. Four clinical (age, histology, stage, and treatment) and 41 18F-FDG-PET-based radiomic features were ranked and a subset of useful features for association with disease progression was selected based on decrease of the Gini impurity. Six machine learning algorithms (random forest, neural network, k-nearest neighbors, naive Bayes, logistic regression, and support vector machine) were compared using the areas under the receiver operating characteristic curve (AUC). Progression-free survival (PFS) was assessed using Cox regression analysis. RESULTS The five top predictors of disease progression were: stage, surface area, metabolic tumor volume, gray-level run length non-uniformity (GLRLM_RLNU), and gray-level non-uniformity for run (GLRLM_GLNU). The naive Bayes model was the best-performing classifier for predicting disease progression (AUC = 0.872, accuracy = 0.780, F1 score = 0.781, precision = 0.788, and recall = 0.780). In the naive Bayes model, 5-year PFS was significantly higher in predicted non-progression than predicted progression (80.1% vs. 9.1%, p < 0.001) and was only the independent factor for PFS in multivariate analysis (HR, 6.89; 95% CI, 1.92-24.69; p = 0.003). CONCLUSION A machine learning approach based on clinical and pretreatment 18F-FDG PET-based radiomic features may be useful for predicting tumor progression in cervical cancer patients.
Collapse
Affiliation(s)
- Masatoyo Nakajo
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan.
| | - Megumi Jinguji
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Atsushi Tani
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Erina Yano
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Chin Khang Hoo
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Daisuke Hirahara
- Department of Management Planning Division, Harada Academy, 2-54-4 Higashitaniyama, Kagoshima, 890-0113, Japan
| | - Shinichi Togami
- Department of Obstetrics and Gynecology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Hiroaki Kobayashi
- Department of Obstetrics and Gynecology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| | - Takashi Yoshiura
- Department of Radiology, Kagoshima University, Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima, 890-8544, Japan
| |
Collapse
|
49
|
Dondi F, Pasinetti N, Gatta R, Albano D, Giubbini R, Bertagna F. Comparison between Two Different Scanners for the Evaluation of the Role of 18F-FDG PET/CT Semiquantitative Parameters and Radiomics Features in the Prediction of Final Diagnosis of Thyroid Incidentalomas. J Clin Med 2022; 11:jcm11030615. [PMID: 35160067 PMCID: PMC8836668 DOI: 10.3390/jcm11030615] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/24/2022] Open
Abstract
The aim of this study was to compare two different tomographs for the evaluation of the role of semiquantitative PET/CT parameters and radiomics features (RF) in the prediction of thyroid incidentalomas (TIs) at 18F-FDG imaging. A total of 221 patients with the presence of TIs were retrospectively included. After volumetric segmentation of each TI, semiquantitative parameters and RF were extracted. All of the features were tested for significant differences between the two PET scanners. The performances of all of the features in predicting the nature of TIs were analyzed by testing three classes of final logistic regression predictive models, one for each tomograph and one with both scanners together. Some RF resulted significantly different between the two scanners. PET/CT semiquantitative parameters were not able to predict the final diagnosis of TIs while GLCM-related RF (in particular GLCM entropy_log2 e GLCM entropy_log10) together with some GLRLM-related and GLZLM-related features presented the best predictive performances. In particular, GLCM entropy_log2, GLCM entropy_log10, GLZLM SZHGE, GLRLM HGRE and GLRLM HGZE resulted the RF with best performances. Our study enabled the selection of some RF able to predict the final nature of TIs discovered at 18F-FDG PET/CT imaging. Classic semiquantitative and volumetric PET/CT parameters did not reveal these abilities. Furthermore, a good overlap in the extraction of RF between the two scanners was underlined.
Collapse
Affiliation(s)
- Francesco Dondi
- Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili Brescia, 25123 Brescia, Italy; (F.D.); (R.G.); (F.B.)
| | - Nadia Pasinetti
- Radiation Oncology Department, ASST Valcamonica Esine and Università degli Studi di Brescia, 25040 Brescia, Italy;
| | - Roberto Gatta
- Dipartimento di Scienze Cliniche e Sperimentali dell’Università degli Studi di Brescia, 25123 Brescia, Italy;
| | - Domenico Albano
- Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili Brescia, 25123 Brescia, Italy; (F.D.); (R.G.); (F.B.)
- Correspondence:
| | - Raffaele Giubbini
- Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili Brescia, 25123 Brescia, Italy; (F.D.); (R.G.); (F.B.)
| | - Francesco Bertagna
- Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili Brescia, 25123 Brescia, Italy; (F.D.); (R.G.); (F.B.)
| |
Collapse
|
50
|
Cheung BMF, Lau KS, Lee VHF, Leung TW, Kong FMS, Luk MY, Yuen KK. Computed tomography-based radiomic model predicts radiological response following stereotactic body radiation therapy in early-stage non-small-cell lung cancer and pulmonary oligo-metastases. Radiat Oncol J 2022; 39:254-264. [PMID: 34986546 PMCID: PMC8743458 DOI: 10.3857/roj.2021.00311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose Radiomic models elaborate geometric and texture features of tumors extracted from imaging to develop predictors for clinical outcomes. Stereotactic body radiation therapy (SBRT) has been increasingly applied in the ablative treatment of thoracic tumors. This study aims to identify predictors of treatment responses in patients affected by early stage non-small cell lung cancer (NSCLC) or pulmonary oligo-metastases treated with SBRT and to develop an accurate machine learning model to predict radiological response to SBRT. Materials and Methods Computed tomography (CT) images of 85 tumors (stage I–II NSCLC and pulmonary oligo-metastases) from 69 patients treated with SBRT were analyzed. Gross tumor volumes (GTV) were contoured on CT images. Patients that achieved complete response (CR) or partial response (PR) were defined as responders. One hundred ten radiomic features were extracted using PyRadiomics module based on the GTV. The association of features with response to SBRT was evaluated. A model using support vector machine (SVM) was then trained to predict response based solely on the extracted radiomics features. Receiver operating characteristic curves were constructed to evaluate model performance of the identified radiomic predictors. Results Sixty-nine patients receiving thoracic SBRT from 2008 to 2018 were retrospectively enrolled. Skewness and root mean squared were identified as radiomic predictors of response to SBRT. The SVM machine learning model developed had an accuracy of 74.8%. The area under curves for CR, PR, and non-responder prediction were 0.86 (95% confidence interval [CI], 0.794–0.921), 0.946 (95% CI, 0.873–0.978), and 0.857 (95% CI, 0.789–0.915), respectively. Conclusion Radiomic analysis of pre-treatment CT scan is a promising tool that can predict tumor response to SBRT.
Collapse
Affiliation(s)
| | - Kin Sang Lau
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong
| | | | - To Wai Leung
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong
| | | | - Mai Yee Luk
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong
| | - Kwok Keung Yuen
- Department of Clinical Oncology, Queen Mary Hospital, Hong Kong
| |
Collapse
|