1
|
Adnan A, Basu S. PET-based Quantitative Techniques in Assessing Efficacy of Interventional Radiology Procedures in Oncology. PET Clin 2025:S1556-8598(25)00028-8. [PMID: 40340172 DOI: 10.1016/j.cpet.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Interventional radiology (IR) is a super specialised branch where imaging modalities are employed to guide disease specific diagnostic and therapeutic interventions. IR interventions have gained popularity in various oncological and non-oncological indications due to it's ability to effectively diagnose the disease and direct specific targeted treatment. Hybrid imaging using PET CT and PET MRI combines the best of morphological and functional informations and offers improved sensitivity and specificity for detection of lesion; helps in accurate mapping of tumour burden, thereby aiding in planning curative vs palliative intent intervention; more accurate response evaluation to plan redo session in cases of residual / recurrent disease or for follow up evaluation and for prognostication and predicting response. Albeit visual analysis of PET images by specialist is most commonly performed for reading PET scans, PET has a remarkable capability to provide quantitative information. The present review provides a comprehensive assessment of the role of various aspects of quantitative PET parameters in assessing the efficacy of IR interventions. The insights provided will help clinicians, researchers, and medical professionals understand the role of PET imaging in advancing patient care and enhancing the therapeutic outcomes of IR procedures.
Collapse
Affiliation(s)
- Aadil Adnan
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, Maharahtra, India; Homi Bhabha National Institute, Mumbai, India; Department of Nuclear Medicine, Medica Cancer Center, Medica Superspeciality Hospital, 127, Eastern Metropolitean Bypass, Mukundapur, Kolkata, West Bengal, India
| | - Sandip Basu
- Radiation Medicine Centre (B.A.R.C), Tata Memorial Centre Annexe, Parel, Mumbai, Maharahtra, India; Homi Bhabha National Institute, Mumbai, India.
| |
Collapse
|
2
|
Kenawy MA, Abdelhafez HM, Al-Fatlawi M, Jassim TN, Jasim AS, Alashkar EM. Comparing texture analysis of pretreatment 18F-FDG PET and 68Ga-PSMA PET in patients with prostate cancer: investigation of diagnostic efficacy and prognostic biomarker. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025; 64:253-261. [PMID: 40137954 DOI: 10.1007/s00411-025-01114-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/12/2025] [Indexed: 03/29/2025]
Abstract
This study aims to evaluate the predictive accuracy of textural parameters and current parameters of 18F-fluorodeoxyglucose and 68Ga-labeled prostate-specific antigen positron emission tomography (FDG and PSMA PET) images in prostate cancer (PCa) and compare the features retrieved from both scans. Based on symptoms, digital rectal examination (DRE), prostate-specific antigen (PSA) level in the blood, or histopathology from transrectal ultrasound-guided biopsy and 4Kscore Test, 120 patients have confirmed PCa. Sixty of them were scanned on a PET/CT machine using 18F-FDG, and the other 60 patients were scanned using 68Ga-PSMA of radiopharmacy. Each tumour was delineated using PET. Edge texture parameters were used to define each tumour, and 73 features in all were taken from eight distinct texture matrices and computed using the open-source program Chang-Gung Image Texture Analysis (CGITA). Using Spearman correlation, feature correlation with conventional quantitative metrics (Maximum Standardized Uptake Value (SUVmax), Total Lesion Glycolysis (TLG), Metabolic Tumor Volume (MTV)) was investigated, and it was found that the High-Intensity Low-Energy Radiation (HILRE) correlation was strong. PCa was best discriminated by HILRE (64-bin) in receiver operating characteristic curves. It is concluded that 68Ga-PSMA-based PET imaging is better than 18F-FDG-based PET and is strongly associated with PCa tumour allocation. According to extracted features, HILRE is the most significant measure and it is, thus, considered here an independent predictor of PCa prognosis. Although the study's findings are helpful, confirmation by further prospective research is required.
Collapse
Affiliation(s)
- Mahmoud A Kenawy
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt.
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq.
| | - Hussein M Abdelhafez
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| | - Murtadha Al-Fatlawi
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Tariq Nadhim Jassim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Ahmed Salman Jasim
- Radiology Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Elsayed M Alashkar
- Biophysics Branch, Department of Physics, Faculty of Science (for Boys), Al-Azhar University, Nasr City, Cairo, 11884, Egypt
| |
Collapse
|
3
|
Zhang W, Zhuang D, Wei W, Yang Y, Ma L, Du H, Jin A, He J, Li X. The 100 most-cited radiomics articles in cancer research: A bibliometric analysis. Clin Imaging 2025; 121:110442. [PMID: 40086035 DOI: 10.1016/j.clinimag.2025.110442] [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: 10/02/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Radiomics, an advanced medical imaging analysis technique introduced by Professor Lambin in 2012, has quickly become a key area of medical research. To explore emerging trends in cancer-related radiomics, we conducted a bibliometric analysis of the 100 most-cited articles (T100) in this field. Data were collected from the Web of Science Core Collection on October 7, 2023, and the articles were ranked by citation count. We extracted data such as authors, journals, citation counts, and publication years and analyzed it using Microsoft Excel 2019 and R 4.4.2. CiteSpace was used to create co-occurrence and citation burst maps to show the relationships between authors, countries, institutions, and keywords. The analysis revealed that the T100 came from 81 countries, with the U.S. contributing the most (56 articles). Harvard University was the leading institution, and the journal Radiology had the highest citation count. Aerts Hugo JWL was the most influential author. The study highlights that "lung cancer" and "artificial intelligence" are emerging as major research hotspots, shaping the future of cancer radiomics.
Collapse
Affiliation(s)
- Wenhao Zhang
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China; Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Dongmei Zhuang
- Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, China
| | - Wenzhuo Wei
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Yuchen Yang
- Department of Clinical Medical, First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Lijun Ma
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - He Du
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Anran Jin
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Jingyi He
- Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China
| | - Xiaoming Li
- Department of Psychiatry, Chaohu Hospital of Anhui Medical University, Hefei, Anhui, China; Department of Medical Psychology, School of Mental Health and Psychological Science, Anhui Medical University, Hefei, China.
| |
Collapse
|
4
|
Liang ZJ, Chang S, Gao Y, Cao W, Kuo LR, Pomeroy MJ, Li LC, Abbasi AF, Bandovic J, Reiter MJ, Pickhardt PJ. Leveraging prior knowledge in machine intelligence to improve lesion diagnosis for early cancer detection. Med Phys 2025. [PMID: 40268724 DOI: 10.1002/mp.17841] [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: 08/17/2024] [Revised: 03/09/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Experts' interpretations of medical images for lesion diagnosis may not always align with the underlying in vivo tissue pathology and, therefore, cannot be considered the definitive truth regarding malignancy or benignity. While current machine learning (ML) models in medical imaging can replicate expert interpretations, their results may also diverge from the actual ground truth. PURPOSE This study investigates various factors contributing to these discrepancies and proposes solutions. METHODS The central idea of the proposed solution is to integrate prior knowledge into ML models to enhance the characterization of in vivo tissues. The incorporation of prior knowledge into decision-making is task-specific, tailored to the data acquired for that task. This central idea was tested on the diagnosis of lesions using low dose computed tomography (LdCT) for early cancer detection, particularly focusing on more challenging, ambiguous or indeterminate lesions (IDLs) as classified by experts. One key piece of prior knowledge involves CT x-ray energy spectrum, where different energies interact with in vivo tissues within a lesion, producing variable but reproducible image contrasts that encapsulate biological information. Typically, CT imaging devices use only the high-energy portion of this spectrum for data acquisition; however, this study considers the full spectrum for lesion diagnostics. Another critical aspect of prior knowledge includes the functional or dynamic properties of in vivo tissues, such as elasticity, which can indicate pathological conditions. Instead of relying solely on abstract image features as current ML models do, this study extracts these tissue pathological characteristics from the image contrast variations. RESULTS The method was tested on LdCT images of four sets of IDLs, including pulmonary nodules and colorectal polyps, with pathological reports serving as the ground truth for malignancy or benignity. The method achieved an area under the receiver operating characteristic curve (AUC) of 0.98 ± 0.03, demonstrating a significant improvement over existing state-of-the-art ML models, which typically have AUCs in the 0.70 range. CONCLUSION Leveraging prior knowledge in machine intelligence can enhance lesion diagnosis, resolve the ambiguity of IDLs interpreted by experts, and improve the effectiveness of LdCT screening for early-stage cancers.
Collapse
Affiliation(s)
- Zhengrong J Liang
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Shaojie Chang
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yongfeng Gao
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
| | - Weiguo Cao
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Licheng R Kuo
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Marc J Pomeroy
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York, USA
| | - Lihong C Li
- Department of Engineering & Environment Science, City University of New York/CSI, Staten Island, New York, USA
| | - Almas F Abbasi
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
| | - Jela Bandovic
- Department of Pathology, Stony Brook University, Stony Brook, New York, USA
| | - Michael J Reiter
- Department of Radiology, Stony Brook University, Stony Brook, New York, USA
| | - Perry J Pickhardt
- Department of Radiology, School of Medicine & Public Health, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
5
|
Gemmell AJ, Brown CM, Ray S, Small A. Robustness of textural analysis features in quantitative 99 mTc and 177Lu SPECT-CT phantom acquisitions. EJNMMI Phys 2025; 12:40. [PMID: 40244535 PMCID: PMC12006590 DOI: 10.1186/s40658-025-00749-0] [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: 07/08/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Textural Analysis features in molecular imaging require to be robust under repeat measurement and to be independent of volume for optimum use in clinical studies. Recent EANM and SNMMI guidelines for radiomics provide advice on the potential use of phantoms to identify robust features (Hatt in EJNMMI, 2022). This study applies the suggested phantoms to use in SPECT quantification for two radionuclides, 99 mTc and 177Lu. METHODS Acquisitions were made with a uniform phantom to test volume dependency and with a customised 'Revolver' phantom, based on the PET phantom described in Hatt (EJNMMI, 2022) but with local adaptations for SPECT. Each phantom was filled separately with 99 mTc and 177Lu. Sixty-seven Textural Analysis features were extracted and tested for robustness and volume dependency. RESULTS Features showing high volume dependency or high Coefficient of Variation (indicating poor repeatability) were removed from the list of features that may be suitable for use in clinical studies. After feature reduction, there were 39 features for 99 mTc and 33 features for 177Lu remaining. CONCLUSION The use of a uniform phantom to test volume dependency and a Revolver phantom to identify repeatable Textural Analysis features is possible for quantitative SPECT using 99 mTc or 177Lu. Selection of such features is likely to be centre-dependent due to differences in camera performance as well as acquisition and reconstruction protocols.
Collapse
Affiliation(s)
- Alastair J Gemmell
- College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK.
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK.
| | - Colin M Brown
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK
| | - Surajit Ray
- School of Mathematics & Statistics, University of Glasgow, Glasgow, UK
| | - Alexander Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow, UK
- Department of Nuclear Medicine, Upper Ground Floor, Gartnavel General Hospital, 1053 Great Western Road, Glasgow, G12 0YN, UK
| |
Collapse
|
6
|
Dwivedi P, Barage S, Jha AK, Choudhury S, Rangarajan V. Robustness of 18F-FDG PET Radiomic Features in Lung Cancer: Impact of Advanced Reconstruction Algorithm. J Nucl Med Technol 2025; 53:50-56. [PMID: 39909582 DOI: 10.2967/jnmt.124.268252] [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: 09/12/2024] [Accepted: 12/02/2024] [Indexed: 02/07/2025] Open
Abstract
18F-FDG PET radiomics is emerging as a promising tool to identify imaging biomarkers for quantifying intratumor heterogeneity in lung cancer. However, the robustness of PET radiomic features (RFs) is influenced by factors such as image reconstruction algorithms, tumor segmentation, and discretization. Although the impact of these factors on RFs has been explored, the specific influence of the advanced block sequential regularized expectation maximization (BSREM) reconstruction algorithm remains unclear. This study investigated the potential variations in PET RFs associated with different factors when using BSREM. Methods: Retrospective data of 18F-FDG PET from 120 lung cancer patients were reconstructed twice using advanced BSREM and conventional ordered-subset expectation maximization methods. For each reconstruction set, 3 tumor segmentations were performed, including manual, 40% threshold, and Nestle methods. Two discretization methods using absolute and relative settings were applied for each dataset before RF extraction. Stable and robust RFs were assessed by the coefficient of variance and intraclass correlation coefficient, respectively. Results: High instability was exhibited by 19%, 33%, and 36% of RFs, with a coefficient of variation of more than 20% for reconstruction, segmentation, and discretization, respectively. Conversely, 60%, 19%, and 35% of RFs demonstrated robustness against these factors, with an intraclass correlation coefficient of more than 0.90. The comparative evaluation revealed significantly greater robustness for most RF subtypes in BSREM than in ordered-subset expectation maximization under varying segmentation and discretization conditions (P < 0.05). Conclusion: The stability and robustness of PET RFs are enhanced if BSREM is applied rather than the conventional method. Study results suggest that the advanced reconstruction method could offer potential benefits in providing consistent PET-based radiomic analysis for improving diagnostic and prognostic value.
Collapse
Affiliation(s)
- Pooja Dwivedi
- Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Amity Institute of Biotechnology, Amity University, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India; and
| | - Sagar Barage
- Amity Institute of Biotechnology, Amity University, Mumbai, India
| | - Ashish Kumar Jha
- Homi Bhabha National Institute, Mumbai, India; and
- Tata Memorial Hospital, Mumbai, India
| | - Sayak Choudhury
- Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India; and
| | - Venkatesh Rangarajan
- Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, India;
- Homi Bhabha National Institute, Mumbai, India; and
- Tata Memorial Hospital, Mumbai, India
| |
Collapse
|
7
|
Feng H, Zhou K, Yuan Q, Liu Z, Zhang T, Chen H, Xu B, Sun Z, Han Z, Liu H, Yu S, Chen T, Li G, Zhou W, Yu J, Huang W, Jiang Y. Noninvasive Assessment of Vascular Endothelial Growth Factor and Prognosis in Gastric Cancer Through Radiomic Features. Clin Transl Gastroenterol 2025; 16:e00802. [PMID: 39787380 PMCID: PMC11932601 DOI: 10.14309/ctg.0000000000000802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 12/04/2024] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Gastric cancer (GC) is a leading cause of cancer-related deaths worldwide, with delayed diagnosis often limiting effective treatment options. This study introduces a novel, noninvasive radiomics-based approach using [18F] FDG PET/CT (fluorodeoxyglucose positron emission tomography/computed tomography) to predict vascular endothelial growth factor (VEGF) status and survival in patients with GC. The ability to noninvasively assess these parameters can significantly influence therapeutic decisions and outcomes. METHODS We conducted a retrospective study involving patients diagnosed with GC, stratified into training, validation, and test groups. Each patient underwent a [18F] FDG PET/CT scan, and radiomic features were extracted using dedicated software. A Radiomics Score (RS) was calculated, serving as a predictor for VEGF status. Statistical analyses included logistic regression and Cox proportional hazards models to evaluate the predictive power of RS on survival outcomes. RESULTS The developed radiomics model demonstrated high predictive accuracy, with the RS formula achieving an area under the receiver operating characteristic curve of 0.861 in the training cohort and 0.857 in the validation cohort for predicting VEGF status. The model also identified RS as an independent prognostic factor for survival, where higher RS values correlated with poorer survival rates. DISCUSSION The findings underscore the potential of [18F] FDG PET/CT radiomics in transforming the management of GC by providing a noninvasive means to assess tumor aggressiveness and prognosis through VEGF status. This model could facilitate earlier and more tailored therapeutic interventions, potentially improving survival outcomes in a disease marked by typically late diagnosis and limited treatment success.
Collapse
Affiliation(s)
- Hao Feng
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kangneng Zhou
- College of Computer Science, Nankai University, Tianjin, China
| | - Qingyu Yuan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhiwei Liu
- Department of PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Taojun Zhang
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Benjamin Xu
- Lynbrook High School, San Jose, California, USA
| | - Zepang Sun
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Han
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Liu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shitong Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Tao Chen
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guoxin Li
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Wenlan Zhou
- Department of PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery & Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weicai Huang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuming Jiang
- Department of Radiation Oncology, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
8
|
Wang Y, Yu Y, Gu L, Sun Y, Yan J, Zhang H, Zhang Y. Radiomics feature is a risk factor for locally advanced cervical cancer treated using concurrent chemoradiotherapy based on magnetic resonance imaging: a retrospective study. BMC Cancer 2025; 25:230. [PMID: 39930343 PMCID: PMC11809009 DOI: 10.1186/s12885-025-13625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Although concurrent chemoradiotherapy (CCRT) is the standard treatment strategy for locally advanced cervical squamous carcinoma (LACSC), there are still individual differences. It is of vital importance to establish a radiomics-based model for predicting overall survival (OS) of LACSC patients treated using CCRT, and evaluating the feasibility of adjuvant chemotherapy (ACT). METHODS 122 LACSC patients were retrospectively analyzed who underwent pelvic MRI before standard CCRT between January 2013 and September 2016, including 85 patients in training set and 37 patients in testing set. 3D Slicer was used to segment images and extract features. IPMs software was used to select features and construct radscore. We selected the group with the largest area under the curves as the best result from 150 feature subsets and corresponding radscore. A nomogram was established using univariate and multivariate Cox analyses. We used Shapley Additive Explanations (SHAP) for further interpretation of the nomogram. Kaplan-Meier curves demonstrated the associations of radscore and clinical characteristics with OS and ACT. RESULTS Radscore was a prognostic factor (P = 0.001) which constructed using 10 radiomics features influencing the OS of patients with LACSC treated using CCRT. The radiomics-clinical model estimated OS (training, C-index: 0.761; testing, C-index: 0.718) more accurately than the clinical (training, C-index: 0.745; testing, C-index: 0.708) and radiomics models (training, C-index: 0.702; testing, C-index: 0.671). Radscore has the greatest impact on the prognosis of LACSC patients. We combined radscore and clinical factors to obtain risk scores. There was a better OS rate among low-risk patients than among high-risk patients (training, P = 0.034; testing, P = 0.003). Compared with CCRT, ACT + CCRT did not improve prognosis (high-risk patients, P = 0.703; all patients, P = 0.425). CONCLUSIONS Radscore independently predicted OS in LACSC. The radiomics-clinical nomogram improved individualized OS estimation. Patients did not benefit from ACT.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yanyan Yu
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lina Gu
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Yunfeng Sun
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiazhuo Yan
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China
| | - Hongxia Zhang
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yunyan Zhang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
9
|
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
|
10
|
Park JE, Oh JY, Park DH, Lee HS, Yoon S, Kim N, Park SY, Song SW, Kim YH, Hong CK, Kim JH, Kim HS. Mapping tumor habitats in isocitrate dehydrogenase -wild type glioblastoma: Integrating MRI, pathologic, and RNA data from the Ivy Glioblastoma Atlas Project. Neuro Oncol 2025; 27:291-301. [PMID: 39177498 PMCID: PMC11726254 DOI: 10.1093/neuonc/noae161] [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: 04/27/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND The goal of the study was to spatially validate intratumoral subregions (tumor habitat) using physiologic magnetic resonance imaging (MRI) on the pathology of the isocitrate dehydrogenase (IDH)-wild-type whole-glioblastoma sample. METHODS Data from 20 patients (168 slides) were obtained from the Ivy Glioblastoma Atlas Project. On MRI, tumor habitats were defined using voxel-wise clustering of the apparent diffusion coefficient and cerebral blood volume maps for contrast-enhancing lesions (CEL) and non-enhancing lesions (NEL). On pathology slides, normalized areas of leading-edge, infiltrating tumor (IT), cellular tumor (CT), hypervascular lesion (CThypervascular), and perinecrotic lesion (CTperinecrotic) were obtained. Gross specimen was coregistered on MRI and correlation between pathology-MRI habitats was calculated. RNA sequencing of 67 samples was assessed using 4 Neftel subtypes and further correlated with pathology. RESULTS Six tumor habitats were identified: hypervascular, hypovascular cellular, and hypovascular hypocellular habitats for CEL and NEL. CT was correlated with hypovascular cellular habitat in CEL (r = 0.238, P = .005). IT was correlated with hypovascular cellular habitat in NEL (r = 0.294, P = .017). CThypervascular was correlated with hypervascular habitat in NEL (r = 0.195, P = .023). CTperinecrotic was correlated with imaging necrosis (r = 0.199, P = .005). Astrocyte-like subtypes were correlated with IT (r = 0.256, P < .001), while mesenchymal-like subtypes were correlated with CTperinecrotic area (r = 0.246, P < .001). CONCLUSIONS Pathologically matched tumor subregions were CT with hypovascular cellular habitat in CEL and infiltrative tumor with hypovascular cellular habitat in NEL. Identification of the most aggressive, as well as infiltrative tumor portion, can be achieved using noninvasive MRI tumor habitats.
Collapse
Affiliation(s)
- Ji Eun Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| | - Joo Young Oh
- Department of Radiation Oncology, Yonsei Cancer Center, Seoul, South Korea
| | - Do Hoon Park
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ho-Su Lee
- Department of Biochemistry and Molecular Biology, Asan Medical Center, Brain Korea 21 Project, University of Ulsan College of Medicine, Seoul, South Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | - Seo Young Park
- Department of Statistics and Data Science, Korea National Open University, Seoul, Korea
| | - Sang Woo Song
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Young-Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Chang-Ki Hong
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Jeong Hoon Kim
- Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Korea
| | - Ho Sung Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, South Korea
| |
Collapse
|
11
|
Sakamoto Y, Yamamoto Y, Uegaki T. [Investigation of the Influence of Image Reconstruction Parameters to Improve the Ability to Depict Internal Tumor Necrosis]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2025; 81:n/a. [PMID: 39864822 DOI: 10.6009/jjrt.25-1453] [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: 01/28/2025]
Abstract
PURPOSE There are attempts to assess tumor heterogeneity by texture analysis. However, the ordered subsets-expectation maximization (OSEM) reconstruction method has problems depicting heterogeneities. The aim of this study was to identify image reconstruction parameters that improve the ability to depict internal tumor necrosis using a self-made phantom that simulates internal necrosis. METHODS Self-made phantoms were prepared using polypropylene cylinders with inner diameters of 18.0 mm and 6.0 mm. The concentration ratios of the simulated tumor : tumor interior were 4 : 0 and 4 : 1. For each reconstruction method, the iteration for OSEM and OSEM+point spread function (PSF) were 1 to 25 and the subset was 12. The β values for block sequential regularized expectation maximization (BSREM) were set between 10 and 400. We evaluated the features of the profile curve, contrast-to-noise ratio, and grey-level co-occurrence matrix (GLCM). RESULTS In the phantom study, OSEM and OSEM+PSF showed a better delineation of the differences between the inside and outside of the cylinder as iteration was increased and BSREM showed a better delineation as β was decreased. The highest value for each feature, both 4 : 0 and 4 : 1, was BSREM β 10 for angular second moment (ASM) and inverse differential moment (IDM), OSEM iteration 25 for contrast and entropy. CONCLUSION We have identified image reconstruction parameters that improve the ability to visualize internal tumor necrosis. The parameter was BRSEM β 10.
Collapse
Affiliation(s)
- Yuka Sakamoto
- Department of Radiology, Nara Prefecture General Medical Center
| | | | - Tadaaki Uegaki
- Department of Radiology, Nara Prefecture General Medical Center
| |
Collapse
|
12
|
Imperiale A, Berti V. SDH-related head and neck paragangliomas: Unraveling PET radiomics beyond 18F-FDG. Best Pract Res Clin Endocrinol Metab 2025; 39:101926. [PMID: 39191631 DOI: 10.1016/j.beem.2024.101926] [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] [Indexed: 08/29/2024]
Abstract
Radiomics revolutionizes medical imaging by providing quantitative analysis that complements traditional qualitative assessments through advanced computational techniques. In this narrative review we have investigated the impact of succinate dehydrogenase (SDH) pathogenic variants on the radiomic profile of 18F-FDG, 18F-DOPA, and 68Ga-DOTA-peptides PET in paragangliomas, focusing on head and neck localizations (HNPGLs). This influence manifests in uptake intensity and textural heterogeneity, revealing a complex radiomic landscape that may reflect specific tumor behaviors and mutation statuses. By combining radiomic analysis with genetic data, we will gain new insights into the relationship between PET imaging features and underlying molecular changes. In the future, we envision an approach integrating macroscopic indices, such as lesion location, size, and SUV, with advanced computer-based algorithms. This comprehensive analysis could facilitate in vivo predictions of SDH pathogenic variants, thereby encouraging genetic testing, and ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, ICANS, University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France; IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France.
| | - Valentina Berti
- Nuclear Medicine Unit, Careggi University Hospital, Florence, Italy; Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', Florence University, Florence, Italy.
| |
Collapse
|
13
|
Zeng S, Wang XL, Yang H. Radiomics and radiogenomics: extracting more information from medical images for the diagnosis and prognostic prediction of ovarian cancer. Mil Med Res 2024; 11:77. [PMID: 39673071 PMCID: PMC11645790 DOI: 10.1186/s40779-024-00580-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies globally. Despite the implementation of various medical imaging approaches for OC screening, achieving accurate differential diagnosis of ovarian tumors continues to pose significant challenges due to variability in image performance, resulting in a lack of objectivity that relies heavily on the expertise of medical professionals. This challenge can be addressed through the emergence and advancement of radiomics, which enables high-throughput extraction of valuable information from conventional medical images. Furthermore, radiomics can integrate with genomics, a novel approach termed radiogenomics, which allows for a more comprehensive, precise, and personalized assessment of tumor biological features. In this review, we present an extensive overview of the application of radiomics and radiogenomics in diagnosing and predicting ovarian tumors. The findings indicate that artificial intelligence methods based on imaging can accurately differentiate between benign and malignant ovarian tumors, as well as classify their subtypes. Moreover, these methods are effective in forecasting survival rates, treatment outcomes, metastasis risk, and recurrence for patients with OC. It is anticipated that these advancements will function as decision-support tools for managing OC while contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Song Zeng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xin-Lu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hua Yang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
14
|
Hinzpeter R, Kulanthaivelu R, Kohan A, Murad V, Mirshahvalad SA, Avery L, Ortega C, Metser U, Hope A, Yeung J, McInnis M, Veit-Haibach P. Predictive [ 18F]-FDG PET/CT-Based Radiogenomics Modelling of Driver Gene Mutations in Non-small Cell Lung Cancer. Acad Radiol 2024; 31:5314-5323. [PMID: 38997880 DOI: 10.1016/j.acra.2024.06.038] [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: 05/07/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024]
Abstract
RATIONALE AND OBJECTIVES To investigate whether [18F]-FDG PET/CT-derived radiomics may correlate with driver gene mutations in non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS In this IRB-approved retrospective study, 203 patients with surgically treated NSCLC who underwent subsequent genomic analysis of the primary tumour at our institution between December 2004 and January 2014 were identified. Of those, 128 patients (mean age 62.4 ± 10.8 years; range: 35-84) received preoperative [18F]-FDG PET/CT as part of their initial staging and thus were included in the study. PET and CT image segmentation and feature extraction were performed semi-automatically with an open-source software platform (LIFEx, Version 6.30, lifexsoft.org). Molecular profiles using different next-generation sequencing (NGS) panels were collected from a web-based resource (cBioPortal.ca for Cancer genomics). Two statistical models were then built to evaluate the predictive ability of [18F]-FDG PET/CT-derived radiomics features for driver gene mutations in NSCLC. RESULTS More than half (68/128, 53%) of all tumour samples harboured three or more gene mutations. Overall, 55% of tumour samples demonstrated a mutation in TP53, 26% of samples had alterations in KRAS and 17% in EGFR. Extensive statistical analysis resulted in moderate to good predictive ability. The highest Youden Index for TP53 was achieved using combined PET/CT features (0.70), for KRAS using PET features only (0.57) and for EGFR using CT features only (0.60). CONCLUSION Our study demonstrated a moderate to good correlation between radiomics features and driver gene mutations in NSCLC, indicating increased predictive ability of genomic profiles using combined [18F]-FDG PET/CT-derived radiomics features.
Collapse
Affiliation(s)
- Ricarda Hinzpeter
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.).
| | - Roshini Kulanthaivelu
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andres Kohan
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Vanessa Murad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Seyed Ali Mirshahvalad
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Lisa Avery
- Department of Biostatistics, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Canada (L.A.); Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada (L.A.)
| | - Claudia Ortega
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Ur Metser
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Andrew Hope
- Department of Radiation Oncology, University Health Network, Toronto, Canada (A.H.)
| | - Jonathan Yeung
- Division of Thoracic Surgery, Department of Surgery, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada (J.Y.)
| | - Micheal McInnis
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| | - Patrick Veit-Haibach
- University Medical Imaging Toronto, Toronto Joint Department Medical Imaging, University Health Network, Sinai Health System, Women's College Hospital, University of Toronto, M5G 2N2, Toronto, Ontario, Canada (R.H., R.K., A.K., V.M., S.A.M., C.O., U.M., M.M., P.V.H.)
| |
Collapse
|
15
|
Kang YK, Ha S, Jeong JB, Oh SW. The value of PET/CT radiomics for predicting survival outcomes in patients with pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:28958. [PMID: 39578496 PMCID: PMC11584753 DOI: 10.1038/s41598-024-77022-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis even without distant metastases, necessitating in-depth characterization of primary tumors for survival prediction. We assessed the feasibility of using FDG-PET/CT radiomics to predict overall survival (OS) in PDAC. This retrospective study included PDAC patients without distant metastasis who underwent FDG-PET/CT for initial staging. Primary tumors were segmented from FDG-PET/CT images, extracting 222 radiomics features. A radiomics-based risk score (Rad-score) was developed using Cox proportional hazards regression with LASSO to predict OS. The prognostic performance of the Rad-score was compared with a clinical model (demographics, disease stage, laboratory results) using Harrell's concordance index (C-index) and bootstrapping. 140 patients were included, with a mortality rate was 72.9% during follow-up (total population, 19.5 ± 19.2 months; survivors, 34.4 ± 28.8 months). Eleven radiomics features were significant for survival prediction. The Rad-score predicted OS with a C-index of 0.681 [95% CI, 0.632-0.731]. A model integrating clinical parameters and Rad-score outperformed the clinical-only model in predicting OS (C-index 0.740 [0.715-0.816] vs. 0.673 [0.650-0.766]; C-index difference 0.067 [0.014-0.113]; P < 0.001). These findings suggest that incorporating FDG-PET/CT radiomics into preexisting prognotic stratification paradiagm may enhance survival prediction in PDAC, warranting large-scale studies to confirm its applicability in clinical practice.
Collapse
Affiliation(s)
- Yeon-Koo Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Ji Bong Jeong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| | - So Won Oh
- Department of Nuclear Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Li C, Lu X, Zhang F, Huang S, Ding L, Wang H, Chen S. Neuroblastoma with high ASPM reveals pronounced heterogeneity and poor prognosis. BMC Cancer 2024; 24:1151. [PMID: 39289658 PMCID: PMC11406734 DOI: 10.1186/s12885-024-12912-4] [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: 11/22/2023] [Accepted: 09/06/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVE We explored the preliminary value of abnormal spindle-like microcephaly- associated (ASPM) protein in aiding precise risk sub-stratification, prediction of metabolic heterogeneity, and prognosis of neuroblastoma (NB). METHODS This retrospective study enrolled newly diagnosed patients with NB who underwent positron emission tomography/computed tomography (PET/CT) before therapy, and tumor tissue was collected after surgery. Regression analysis was used to evaluate ASPM expression and risk stratification in patients with NB. The expression levels of ASPM, clinical information, and PET/CT text features were analyzed using univariate and multivariate survival analyses. Finally, a correlation analysis was used to explore the relationship between ASPM and tumor metabolic heterogeneity. RESULTS There were 48 patients with NB in this study (35 boys and 13 girls); 22 patients progressed and 16 died. We found that the level of ASPM was highly associated with risk stratification (OR = 5.295, 95%IC: 1.348-41.722, p = 0.021). Patients with NB and high-risk stratification with high ASPM level had a lower 3-year progression-free survival (PFS) rate (14.28%) and 1-year PFS rate (57.14%) than those with low ASPM level (57.14% and 93.75%, respectively). Using univariate and multivariate survival analyses, this study revealed that ASPM and LDH were independent risk factors for both PFS and overall survival (OS), whales GLZLM_ZLNU was only a risk factor for PFS. CONCLUSION ASPM holds promise as a novel biomarker for refining current risk stratification and predicting prognosis in neuroblastoma. Elevated levels of ASPM, LDH, and GLZLM_ZLNU may be associated with poorer survival outcomes in neuroblastoma patients.
Collapse
Affiliation(s)
- Chao Li
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xueyuan Lu
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Fengxian Zhang
- Department of Nuclear Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, 507 Zhengmin Road, Shanghai, 200433, China
| | - Shuo Huang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Lin Ding
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Hui Wang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Suyun Chen
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
17
|
Pellegrino S, Origlia D, Di Donna E, Lamagna M, Della Pepa R, Pane F, Del Vecchio S, Fonti R. Coefficient of variation and texture analysis of 18F-FDG PET/CT images for the prediction of outcome in patients with multiple myeloma. Ann Hematol 2024; 103:3713-3721. [PMID: 39046513 PMCID: PMC11358233 DOI: 10.1007/s00277-024-05905-7] [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: 02/28/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
In multiple myeloma (MM) bone marrow infiltration by monoclonal plasma cells can occur in both focal and diffuse manner, making staging and prognosis rather difficult. The aim of our study was to test whether texture analysis of 18 F-2-deoxy-d-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) images can predict survival in MM patients. Forty-six patients underwent 18 F-FDG-PET/CT before treatment. We used an automated contouring program for segmenting the hottest focal lesion (FL) and a lumbar vertebra for assessing diffuse bone marrow involvement (DI). Maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean) and texture features such as Coefficient of variation (CoV), were obtained from 46 FL and 46 DI. After a mean follow-up of 51 months, 24 patients died of myeloma and were compared to the 22 survivors. At univariate analysis, FL SUVmax (p = 0.0453), FL SUVmean (p = 0.0463), FL CoV (p = 0.0211) and DI SUVmax (p = 0.0538) predicted overall survival (OS). At multivariate analysis only FL CoV and DI SUVmax were retained in the model (p = 0.0154). By Kaplan-Meier method and log-rank testing, patients with FL CoV below the cut-off had significantly better OS than those with FL CoV above the cut-off (p = 0.0003), as well as patients with DI SUVmax below the threshold versus those with DI SUVmax above the threshold (p = 0.0006). Combining FL CoV and DI SUVmax by using their respective cut-off values, a statistically significant difference was found between the resulting four survival curves (p = 0.0001). Indeed, patients with both FL CoV and DI SUVmax below their respective cut-off values showed the best prognosis. Conventional and texture parameters derived from 18F-FDG PET/CT analysis can predict survival in MM patients by assessing the heterogeneity and aggressiveness of both focal and diffuse infiltration.
Collapse
Affiliation(s)
- Sara Pellegrino
- Department of Advanced Biomedical Sciences, University Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Davide Origlia
- Department of Advanced Biomedical Sciences, University Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Erica Di Donna
- Department of Advanced Biomedical Sciences, University Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Martina Lamagna
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Roberta Della Pepa
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, University Federico II, Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University Federico II, Via Sergio Pansini 5, Naples, 80131, Italy
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University Federico II, Via Sergio Pansini 5, Naples, 80131, Italy.
| |
Collapse
|
18
|
Ai Y, Zhu X, Zhang Y, Li W, Li H, Zhao Z, Zhang J, Ning B, Li C, Zheng Q, Zhang J, Jin J, Li Y, Xie C, Jin X. MRI radiomics nomogram integrating postoperative adjuvant treatments in recurrence risk prediction for patients with early-stage cervical cancer. Radiother Oncol 2024; 197:110328. [PMID: 38761884 DOI: 10.1016/j.radonc.2024.110328] [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: 10/07/2023] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND AND PURPOSE Adjuvant treatments are valuable to decrease the recurrence rate and improve survival for early-stage cervical cancer patients (ESCC), Therefore, recurrence risk evaluation is critical for the choice of postoperative treatment. A magnetic resonance imaging (MRI) based radiomics nomogram integrating postoperative adjuvant treatments was constructed and validated externally to improve the recurrence risk prediction for ESCC. MATERIAL AND METHODS 212 ESCC patients underwent surgery and adjuvant treatments from three centers were enrolled and divided into the training, internal validation, and external validation cohorts. Their clinical data, pretreatment T2-weighted images (T2WI) were retrieved and analyzed. Radiomics models were constructed using machine learning methods with features extracted and screen from sagittal and axial T2WI. A nomogram for recurrence prediction was build and evaluated using multivariable logistic regression analysis integrating radiomic signature and adjuvant treatments. RESULTS A total of 8 radiomic features were screened out of 1020 extracted features. The extreme gradient boosting (XGboost) model based on MRI radiomic features performed best in recurrence prediction with an area under curve (AUC) of 0.833, 0.822 in the internal and external validation cohorts, respectively. The nomogram integrating radiomic signature and clinical factors achieved an AUC of 0.806, 0.718 in the internal and external validation cohorts, respectively, for recurrence risk prediction for ESCC. CONCLUSION In this study, the nomogram integrating T2WI radiomic signature and clinical factors is valuable to predict the recurrence risk, thereby allowing timely planning for effective treatments for ESCC with high risk of recurrence.
Collapse
Affiliation(s)
- Yao Ai
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoyang Zhu
- Department of Radiotherapy, the Second Affiliated Hospital Zhejiang University School of Medicine, Zhejiang, China
| | - Yu Zhang
- Department of Information Division, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenlong Li
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Heng Li
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zeshuo Zhao
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jicheng Zhang
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Boda Ning
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chenyu Li
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiao Zheng
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ji Zhang
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Juebin Jin
- Department of Medical Engineering, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yiran Li
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Xiance Jin
- Radiotherapy Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China; School of Basic Medical Science, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
19
|
Chao F, Wang R, Han X, Huang W, Wang R, Yu Y, Lin X, Yuan P, Yang M, Gao J. Intratumoral metabolic heterogeneity by 18F-FDG PET/CT to predict prognosis for patients with thymic epithelial tumors. Thorac Cancer 2024; 15:1437-1445. [PMID: 38757212 PMCID: PMC11194121 DOI: 10.1111/1759-7714.15331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND The aim of the present study was to evaluate the impact of intratumoral metabolic heterogeneity and quantitative 18F-FDG PET/CT imaging parameters in predicting patient outcomes in thymic epithelial tumors (TETs). METHODS This retrospective study included 100 patients diagnosed with TETs who underwent pretreatment 18F-FDG PET/CT. The maximum and mean standardized uptake values (SUVmax and SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) on PET/CT were measured. Heterogeneity index-1 (HI-1; standard deviation [SD] divided by SUVmean) and heterogeneity index-2 (HI-2; linear regression slopes of the MTV according with different SUV thresholds), were evaluated as heterogeneity indices. Associations between these parameters and patient survival outcomes were analyzed. RESULTS The univariate analysis showed that Masaoka stage, TNM stage, WHO classification, SUVmax, SUVmean, TLG, and HI-1 were significant prognostic factors for progression-free survival (PFS), while MTV, HI-2, age, gender, presence of myasthenia gravis, and maximum tumor diameter were not. Subsequently, multivariate analyses showed that HI-1 (p < 0.001) and TNM stage (p = 0.002) were independent prognostic factors for PFS. For the overall survival analysis, TNM stage, WHO classification, SUVmax, and HI-1 were significant prognostic factors in the univariate analysis, while TNM stage remained an independent prognostic factor in multivariate analyses (p = 0.024). The Kaplan Meier survival analyses showed worse prognoses for patients with TNM stages III and IV and HI-1 ≥ 0.16 compared to those with stages I and II and HI-1 < 0.16 (log-rank p < 0.001). CONCLUSION HI-1 and TNM stage were independent prognostic factors for progression-free survival in TETs. HI-1 generated from baseline 18F-FDG PET/CT might be promising to identify patients with poor prognosis.
Collapse
Affiliation(s)
- Fangfang Chao
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ran Wang
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xingmin Han
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Wenpeng Huang
- Department of Nuclear MedicinePeking University First HospitalBeijingChina
| | - Ruihua Wang
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yanxia Yu
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xuyang Lin
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ping Yuan
- Department of Thoracic SurgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Meng Yang
- Department of Nuclear MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Jianbo Gao
- Department of RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
20
|
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
|
21
|
Wang G, Si Y, Liu J, Wang W, Yang J. Prognostic Value of Metabolic Parameters and Textural Features in Pretreatment 18F-FDG PET/CT of Primary Lesions for Pediatric Patients with Neuroblastoma. Acad Radiol 2024; 31:1091-1101. [PMID: 37748956 DOI: 10.1016/j.acra.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/05/2023] [Accepted: 08/07/2023] [Indexed: 09/27/2023]
Abstract
RATIONALE AND OBJECTIVES Our study evaluated the prognostic value of the metabolic parameters and textural features in pretreatment 18F-Fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) of primary lesions for pediatric patients with neuroblastoma. MATERIALS AND METHODS In total, 107 pediatric patients with neuroblastoma who underwent pretreatment 18F-FDG PET/CT were retrospectively included and analyzed. All patients were diagnosed by pathology, and baseline characteristics and clinical data were collected. The four metabolic parameters and 43 textural features of 18F-FDG PET/CT of the primary lesions were measured. The prognostic significance of metabolic parameters and other clinical variables was assessed using Cox proportional hazards regression models. Differences in progression-free survival (PFS) and overall survival (OS) in relation to parameters were examined using the Kaplan-Meier method. RESULTS During a median follow-up period of 34.3 months, 45 patients (42.1%) experienced tumor recurrence or progression, and 21 patients (19.6%) died of cancer. In univariate Cox regression analysis, age, location of disease, International Neuroblastoma Risk Group Staging System (INRGSS) stage M, neuron-specific enolase (NSE), lactate dehydrogenase (LDH), four positron emission tomography (PET) metabolic parameters, and 33 textural features were significant predictors of PFS. In multivariate analysis, INRGSS stage M (hazard ratio [HR] = 19.940, 95% confidence interval [CI] = 2.733-145.491, P = 0.003), skewness (>0.173; PET first-order features; HR = 2.938, 95% CI = 1.389-6.215, P = 0.005), coarseness (>0.003; neighborhood gray-tone difference matrix; HR = 0.253, 95% CI = 0.132-0.484, P < 0.001), and variance (>103.837; CT first-order gray histogram parameters; HR = 2.810, 95% CI = 1.160-6.807, P = 0.022) were independent predictors of PFS. In univariate Cox regression analysis, gender, INRGSS stage M, MYCN amplification, NSE, LDH, two PET metabolic parameters, and five textural features were significant predictors of OS. In multivariate analysis, INRGSS stage M (HR = 7.704, 95% CI = 1.031-57.576, P = 0.047), MYCN amplification (HR = 3.011, 95% CI = 1.164-7.786, P = 0.023), and metabolic tumor volume (>138.788; HR = 3.930, 95% CI = 1.317-11.727, P = 0.014) were independent predictors of OS. CONCLUSION The metabolic parameters and textural features in pretreatment 18F-FDG PET/CT of primary lesions are predictive of survival in pediatric patients with neuroblastoma.
Collapse
Affiliation(s)
- Guanyun Wang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China (G.W., J.L., W.W., J.Y.)
| | - Yukun Si
- UItrasonic Diagnosis Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing, China, 100050 (Y.S.)
| | - Jun Liu
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China (G.W., J.L., W.W., J.Y.)
| | - Wei Wang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China (G.W., J.L., W.W., J.Y.)
| | - Jigang Yang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China (G.W., J.L., W.W., J.Y.).
| |
Collapse
|
22
|
Feng L, Yao X, Lu X, Wang C, Wang W, Yang J. Differentiation of early relapse and late relapse in intermediate- and high-risk neuroblastoma with an 18F-FDG PET/CT-based radiomics nomogram. Abdom Radiol (NY) 2024; 49:888-899. [PMID: 38315193 DOI: 10.1007/s00261-023-04181-9] [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: 11/18/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 02/07/2024]
Abstract
OBJECTIVES To develop and validate an 18F-FDG PET/CT-based radiomics nomogram for differentiating early relapse and late relapse of intermediate- and high-risk neuroblastoma (NB). METHODS A total of eighty-five patients with relapsed NB who underwent 18F-FDG PET/CT were retrospectively evaluated. All selected patients were randomly assigned to the training set and the validation set in a 7:3 ratio. Tumors were segmented using the 3D slicer, followed by radiomics features extraction. Features selection was performed using random forest, and the radiomics score was constructed by logistic regression analysis. Clinical risk factors were identified, and the clinical model was constructed using logistic regression analysis. A radiomics nomogram was constructed by combining the radiomics score and clinical risk factors, and its performance was evaluated by receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). RESULTS Finally, the 12 most important radiomics features were used for modeling, with an area under the curve (AUC) of 0.835 and 0.824 in the training and validation sets, respectively. Age at diagnosis and International Neuroblastoma Pathology Classification were determined as clinical risk factors to construct the clinical model. In addition, the nomogram achieved an AUC of 0.902 and 0.889 for identifying early relapse in the training and validation sets, respectively, which is higher than the clinical model (AUC of 0.712 and 0.588, respectively). The predicted early relapse and actual early relapse in the calibration curves were in good agreement. The DCA showed that the radiomics nomogram was clinically useful. CONCLUSION Our 18F-FDG PET/CT-based radiomics nomogram can well predict early relapse and late relapse of intermediate- and high-risk NB, which contributes to follow-up and management in clinical practice.
Collapse
Affiliation(s)
- Lijuan Feng
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xilan Yao
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Xia Lu
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Chao Wang
- SinoUnion Healthcare Inc., Beijing, China
| | - Wei Wang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China
| | - Jigang Yang
- Department of Nuclear Medicine, Beijing Friendship Hospital, Capital Medical University, 95 Yong An Road, Xi Cheng District, Beijing, China.
| |
Collapse
|
23
|
Choi W, Jia Y, Kwak J, Werner-Wasik M, Dicker AP, Simone NL, Storozynsky E, Jain V, Vinogradskiy Y. Novel Functional Radiomics for Prediction of Cardiac Positron Emission Tomography Avidity in Lung Cancer Radiotherapy. JCO Clin Cancer Inform 2024; 8:e2300241. [PMID: 38452302 PMCID: PMC10939651 DOI: 10.1200/cci.23.00241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/22/2023] [Accepted: 01/26/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE Traditional methods of evaluating cardiotoxicity focus on radiation doses to the heart. Functional imaging has the potential to provide improved prediction for cardiotoxicity for patients with lung cancer. Fluorine-18 (18F) fluorodeoxyglucose (FDG)-positron emission tomography (PET)/computed tomography (CT) imaging is routinely obtained in a standard cancer staging workup. This work aimed to develop a radiomics model predicting clinical cardiac assessment using 18F-FDG PET/CT scans before thoracic radiation therapy. METHODS Pretreatment 18F-FDG PET/CT scans from three study populations (N = 100, N = 39, N = 70) were used, comprising two single-institutional protocols and one publicly available data set. A clinician (V.J.) classified the PET/CT scans per clinical cardiac guidelines as no uptake, diffuse uptake, or focal uptake. The heart was delineated, and 210 novel functional radiomics features were selected to classify cardiac FDG uptake patterns. Training data were divided into training (80%)/validation (20%) sets. Feature reduction was performed using the Wilcoxon test, hierarchical clustering, and recursive feature elimination. Ten-fold cross-validation was carried out for training, and the accuracy of the models to predict clinical cardiac assessment was reported. RESULTS From 202 of 209 scans, cardiac FDG uptake was scored as no uptake (39.6%), diffuse uptake (25.3%), and focal uptake (35.1%), respectively. Sixty-two independent radiomics features were reduced to nine clinically pertinent features. The best model showed 93% predictive accuracy in the training data set and 80% and 92% predictive accuracy in two external validation data sets. CONCLUSION This work used an extensive patient data set to develop a functional cardiac radiomic model from standard-of-care 18F-FDG PET/CT scans, showing good predictive accuracy. The radiomics model has the potential to provide an automated method to predict existing cardiac conditions and provide an early functional biomarker to identify patients at risk of developing cardiac complications after radiotherapy.
Collapse
Affiliation(s)
- Wookjin Choi
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Yingcui Jia
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Jennifer Kwak
- Department of Radiology, University of Colorado School of Medicine, Aurora, CO
| | - Maria Werner-Wasik
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Adam P. Dicker
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Nicole L. Simone
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Eugene Storozynsky
- Department of Cardiology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Varsha Jain
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | - Yevgeniy Vinogradskiy
- Department of Radiation Oncology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
24
|
Zheng X, Huang Y, Lin Y, Zhu T, Zou J, Wang S, Wang K. 18F-FDG PET/CT-based deep learning radiomics predicts 5-years disease-free survival after failure to achieve pathologic complete response to neoadjuvant chemotherapy in breast cancer. EJNMMI Res 2023; 13:105. [PMID: 38052965 DOI: 10.1186/s13550-023-01053-7] [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: 08/02/2023] [Accepted: 11/19/2023] [Indexed: 12/07/2023] Open
Abstract
BACKGROUND This study aimed to assess whether a combined model incorporating radiomic and depth features extracted from PET/CT can predict disease-free survival (DFS) in patients who failed to achieve pathologic complete response (pCR) after neoadjuvant chemotherapy. RESULTS This study retrospectively included one hundred and five non-pCR patients. After a median follow-up of 71 months, 15 and 7 patients experienced recurrence and death, respectively. The primary tumor volume underwent feature extraction, yielding a total of 3644 radiomic features and 4096 depth features. The modeling procedure employed Cox regression for feature selection and utilized Cox proportional-hazards models to make predictions on DFS. Time-dependent receiver operating characteristic (ROC) curves and the area under the ROC curve (AUC) were utilized to evaluate and compare the predictive performance of different models. 2 clinical features (RCB, cT), 4 radiomic features, and 7 depth features were significant predictors of DFS and were included to develop models. The integrated model incorporating RCB, cT, and radiomic and depth features extracted from PET/CT images exhibited the highest accuracy for predicting 5-year DFS in the training (AUC 0.943) and the validation cohort (AUC 0.938). CONCLUSION The integrated model combining radiomic and depth features extracted from PET/CT images can accurately predict 5-year DFS in non-pCR patients. It can help identify patients with a high risk of recurrence and strengthen adjuvant therapy to improve survival.
Collapse
Affiliation(s)
- Xingxing Zheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yuhong Huang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yingyi Lin
- Shantou University Medical College, Shantou, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiachen Zou
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Medical University, Zhanjiang, China
| | - Shuxia Wang
- Department of Nuclear Medicine and PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
25
|
Lai Q, Zhao Y, Yan H, Peng H. Advances in diagnosis, treatment and prognostic factors of gastrointestinal DLBCL. Leuk Res 2023; 135:107406. [PMID: 37944240 DOI: 10.1016/j.leukres.2023.107406] [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: 08/12/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023]
Abstract
Gastrointestinal diffuse large B-cell lymphoma (GI-DLBCL) is an extremely aggressive form of B-cell non-Hodgkin lymphoma (BNHL) which has complex histological characteristics and manifests a high degree of heterogeneity in terms of clinical, morphological, immunological, and genetic features. GI-DLBCL mainly spreads by infiltrating neighboring lymph nodes, and common gastrointestinal complications (GICS) such as obstruction, perforation, or bleeding, frequently arise during the progression of the disease, posing significant challenges in both diagnosing and treating the condition. Meanwhile, the incidence of GI-DLBCL has been gradually increasing in recent years, and its strong invasiveness makes it prone to being misdiagnosed or completely missed. In clinical practice, over half of the patients diagnosed with the disease are in stage III or stage IV. What makes it worse is that certain patients may not exhibit a favorable response to chemotherapy. All these lead to intricacies in management of this disease. Unfortunately, there is currently no large prospective study or evidence-based medical evidence to provide clear guidance on treatment decisions for this specific type of lymphoma. Neither do physicians have a consensus regarding the optimal approach to address this condition. Recent studies have identified the presence of various prognostic factors that significantly impact survival in GI-DLBCL, which demonstrates the unique particularity of GI-DLBCL, and could help optimize the clinical decision.
Collapse
Affiliation(s)
- Qinqiao Lai
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiqing Yan
- Department of gastric and abdominal cancer ward, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Engineering Research Center of Cell Immunotherapy for Hematopoietic Malignancies, Changsha, Hunan, China.
| |
Collapse
|
26
|
Ning J, Li C, Yu P, Cui J, Xu X, Jia Y, Zuo P, Tian J, Kenner L, Xu B. Radiomic analysis will add differential diagnostic value of benign and malignant pulmonary nodules: a hybrid imaging study based on [ 18F]FDG and [ 18F]FLT PET/CT. Insights Imaging 2023; 14:197. [PMID: 37980611 PMCID: PMC10657912 DOI: 10.1186/s13244-023-01530-6] [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: 12/31/2022] [Accepted: 09/25/2023] [Indexed: 11/21/2023] Open
Abstract
PURPOSE To investigate the clinical value of radiomic analysis on [18F]FDG and [18F]FLT PET on the differentiation of [18F]FDG-avid benign and malignant pulmonary nodules (PNs). METHODS Data of 113 patients with inconclusive PNs based on preoperative [18F]FDG PET/CT who underwent additional [18F]FLT PET/CT scans within a week were retrospectively analyzed in the present study. Three methods of analysis including visual analysis, radiomic analysis based on [18F]FDG PET/CT images alone, and radiomic analysis based on dual-tracer PET/CT images were evaluated for differential diagnostic value of benign and malignant PNs. RESULTS A total of 678 radiomic features were extracted from volumes of interest (VOIs) of 123 PNs. Fourteen valuable features were thereafter selected. Based on a visual analysis of [18F]FDG PET/CT images, the diagnostic accuracy, sensitivity, and specificity were 61.6%, 90%, and 28.8%, respectively. For the test set, the area under the curve (AUC), sensitivity, and specificity of the radiomic models based on [18F]FDG PET/CT plus [18F]FLT signature were equal or better than radiomics based on [18F]FDG PET/CT only (0.838 vs 0.810, 0.778 vs 0.778, 0.750 vs 0.688, respectively). CONCLUSION Radiomic analysis based on dual-tracer PET/CT images is clinically promising and feasible for the differentiation between benign and malignant PNs. CLINICAL RELEVANCE STATEMENT Radiomic analysis will add differential diagnostic value of benign and malignant pulmonary nodules: a hybrid imaging study based on [18F]FDG and [18F]FLT PET/CT. KEY POINTS • Radiomics brings new insights into the differentiation of benign and malignant pulmonary nodules beyond the naked eyes. • Dual-tracer imaging shows the biological behaviors of cancerous cells from different aspects. • Radiomics helps us get to the histological view in a non-invasive approach.
Collapse
Affiliation(s)
- Jing Ning
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Vienna General Hospital, Vienna, Austria
- Department of Clinical Pathology, Vienna General Hospital, Vienna, Austria
| | - Can Li
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng Yu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jingjing Cui
- United Imaging Intelligence (Beijing) Co., Ltd., Beijing, China Yongteng North Road, Haidian District, Beijing, China
| | - Xiaodan Xu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Jia
- Huiying Medical Technology Co., Ltd., Room C103, B2, Dongsheng Science and Technology Park, Haidian District, Beijing, China
| | - Panli Zuo
- Huiying Medical Technology Co., Ltd., Room C103, B2, Dongsheng Science and Technology Park, Haidian District, Beijing, China
| | - Jiahe Tian
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lukas Kenner
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Vienna, Austria.
| | - Baixuan Xu
- Department of Nuclear Medicine, First Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
27
|
Travaglio Morales D, Huerga Cabrerizo C, Losantos García I, Coronado Poggio M, Cordero García JM, Llobet EL, Monachello Araujo D, Rizkallal Monzón S, Domínguez Gadea L. Prognostic 18F-FDG Radiomic Features in Advanced High-Grade Serous Ovarian Cancer. Diagnostics (Basel) 2023; 13:3394. [PMID: 37998530 PMCID: PMC10670627 DOI: 10.3390/diagnostics13223394] [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: 09/27/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/25/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is an aggressive disease with different clinical outcomes and poor prognosis. This could be due to tumor heterogeneity. The 18F-FDG PET radiomic parameters permit addressing tumor heterogeneity. Nevertheless, this has been not well studied in ovarian cancer. The aim of our work was to assess the prognostic value of pretreatment 18F-FDG PET radiomic features in patients with HGSOC. A review of 36 patients diagnosed with advanced HGSOC between 2016 and 2020 in our center was performed. Radiomic features were obtained from pretreatment 18F-FDGPET. Disease-free survival (DFS) and overall survival (OS) were calculated. Optimal cutoff values with receiver operating characteristic curve/median values were used. A correlation between radiomic features and DFS/OS was made. The mean DFS was 19.6 months and OS was 37.1 months. Total Lesion Glycolysis (TLG), GLSZM_ Zone Size Non-Uniformity (GLSZM_ZSNU), and GLRLM_Run Length Non-Uniformity (GLRLM_RLNU) were significantly associated with DFS. The survival-curves analysis showed a significant difference of DSF in patients with GLRLM_RLNU > 7388.3 versus patients with lower values (19.7 months vs. 31.7 months, p = 0.035), maintaining signification in the multivariate analysis (p = 0.048). Moreover, Intensity-based Kurtosis was associated with OS (p = 0.027). Pretreatment 18F-FDG PET radiomic features GLRLM_RLNU, GLSZM_ZSNU, and Kurtosis may have prognostic value in patients with advanced HGSOC.
Collapse
Affiliation(s)
- Daniela Travaglio Morales
- Nuclear Medicine Department, La Paz University Hospital, 28046 Madrid, Spain
- Nuclear Medicine Department, Halle University Hospital, 06120 Halle, Germany
| | - Carlos Huerga Cabrerizo
- Department of Medical Physics and Radiation Protection, La Paz University Hospital, 28046 Madrid, Spain
| | | | | | | | - Elena López Llobet
- Nuclear Medicine Department, La Paz University Hospital, 28046 Madrid, Spain
| | | | | | | |
Collapse
|
28
|
Yoon H, Choi WH, Joo MW, Ha S, Chung YA. SPECT/CT Radiomics for Differentiating between Enchondroma and Grade I Chondrosarcoma. Tomography 2023; 9:1868-1875. [PMID: 37888740 PMCID: PMC10610631 DOI: 10.3390/tomography9050148] [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: 08/30/2023] [Revised: 10/11/2023] [Accepted: 10/14/2023] [Indexed: 10/28/2023] Open
Abstract
This study was performed to assess the value of SPECT/CT radiomics parameters in differentiating enchondroma and atypical cartilaginous tumors (ACTs) located in the long bones. Quantitative HDP SPECT/CT data of 49 patients with enchondromas or ACTs in the long bones were retrospectively reviewed. Patients were randomly split into training (n = 32) and test (n = 17) data, and SPECT/CT radiomics parameters were extracted. In training data, LASSO was employed for feature reduction. Selected parameters were compared with classic quantitative parameters for the prediction of diagnosis. Significant parameters from training data were again tested in the test data. A total of 12 (37.5%) and 6 (35.2%) patients were diagnosed as ACTs in training and test data, respectively. LASSO regression selected two radiomics features, zone-length non-uniformity for zone (ZLNUGLZLM) and coarseness for neighborhood grey-level difference (CoarsenessNGLDM). Multivariate analysis revealed higher ZLNUGLZLM as the only significant independent factor for the prediction of ACTs, with sensitivity and specificity of 85.0% and 58.3%, respectively, with a cut-off value of 191.26. In test data, higher ZLNUGLZLM was again associated with the diagnosis of ACTs, with sensitivity and specificity of 83.3% and 90.9%, respectively. HDP SPECT/CT radiomics may provide added value for differentiating between enchondromas and ACTs.
Collapse
Affiliation(s)
- Hyukjin Yoon
- Division of Nuclear Medicine, Department of Radiology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.Y.); (W.H.C.)
| | - Woo Hee Choi
- Division of Nuclear Medicine, Department of Radiology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (H.Y.); (W.H.C.)
| | - Min Wook Joo
- Department of Orthopedic Surgery, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Yong-An Chung
- Division of Nuclear Medicine, Department of Radiology, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
29
|
Zheng X, Huang C, Yu B, Liu S, Li T, Guan Y, Ding J. Differentiation of neurogenic tumours and pleomorphic adenomas in the parapharyngeal space based on texture analysis of T2WI. BMC Oral Health 2023; 23:548. [PMID: 37559074 PMCID: PMC10413588 DOI: 10.1186/s12903-023-03283-6] [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: 09/03/2022] [Accepted: 08/03/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND The purpose of this study was to identify neurogenic tumours and pleomorphic adenomas of the parapharyngeal space based on the texture characteristics of MRI-T2WI. METHODS MR findings and pathological reports of 25 patients with benign tumours in the parapharyngeal space were reviewed retrospectively (13 cases with pleomorphic adenomas and 12 cases with neurogenic tumours). Using PyRadiomics, the texture of the region of interest in T2WI sketched by radiologists was analysed. By using independent sample t-tests and Mann‒Whitney U tests, the selected texture features of 36 Gray Level Co-Occurrence Matrix (GLCM) and Gray Level Dependence Matrix (GLDM) were tested. A set of parameters of texture features showed statistically significant differences between the two groups, which were selected, and the diagnostic efficiency was evaluated via the operating characteristic curve of the subjects. RESULTS The differences in the three parameters - small dependence low level emphasis (SDLGLE), low level emphasis (LGLE) and difference variance (DV) of characteristics - between the two groups were statistically significant (P < 0.05). No significant difference was found in the other indices. ROC curves were drawn for the three parameters, with AUCs of 0.833, 0.795, and 0.744, respectively. CONCLUSIONS There is a difference in the texture characteristic parameters based on magnetic resonance T2WI images between neurogenic tumours and pleomorphic adenomas in the parapharyngeal space. For the differential diagnosis of these two kinds of tumours, texture analysis of significant importance is an objective and quantitative analytical tool.
Collapse
Affiliation(s)
- Xuewei Zheng
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China
| | - Chencui Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise and League of PHD Technology Co. Ltd., Beijing, 100080, China
| | - Baoting Yu
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China
| | - Shuo Liu
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China
| | - Tong Li
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China
| | - Yuyao Guan
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China
| | - Jun Ding
- Department of Radiology, China-Japan Union Hospital of Jilin University, No. 829 of Xinmin Street, Chaoyang District, Changchun, 130021, China.
| |
Collapse
|
30
|
Philip MM, Welch A, McKiddie F, Nath M. A systematic review and meta-analysis of predictive and prognostic models for outcome prediction using positron emission tomography radiomics in head and neck squamous cell carcinoma patients. Cancer Med 2023; 12:16181-16194. [PMID: 37353996 PMCID: PMC10469753 DOI: 10.1002/cam4.6278] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/07/2023] [Accepted: 06/11/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Positron emission tomography (PET) images of head and neck squamous cell carcinoma (HNSCC) patients can assess the functional and biochemical processes at cellular levels. Therefore, PET radiomics-based prediction and prognostic models have the potentials to understand tumour heterogeneity and assist clinicians with diagnosis, prognosis and management of the disease. We conducted a systematic review of published modelling information to evaluate the usefulness of PET radiomics in the prediction and prognosis of HNSCC patients. METHODS We searched bibliographic databases (MEDLINE, Embase, Web of Science) from 2010 to 2021 and considered 31 studies with pre-defined inclusion criteria. We followed the CHARMS checklist for data extraction and performed quality assessment using the PROBAST tool. We conducted a meta-analysis to estimate the accuracy of the prediction and prognostic models using the diagnostic odds ratio (DOR) and average C-statistic, respectively. RESULTS Manual segmentation method followed by 40% of the maximum standardised uptake value (SUVmax ) thresholding is a commonly used approach. The area under the receiver operating curves of externally validated prediction models ranged between 0.60-0.87, 0.65-0.86 and 0.62-0.75 for overall survival, distant metastasis and recurrence, respectively. Most studies highlighted an overall high risk of bias (outcome definition, statistical methodologies and external validation of models) and high unclear concern in terms of applicability. The meta-analysis showed the estimated pooled DOR of 6.75 (95% CI: 4.45, 10.23) for prediction models and the C-statistic of 0.71 (95% CI: 0.67, 0.74) for prognostic models. CONCLUSIONS Both prediction and prognostic models using clinical variables and PET radiomics demonstrated reliable accuracy for detecting adverse outcomes in HNSCC, suggesting the prospect of PET radiomics in clinical settings for diagnosis, prognosis and management of HNSCC patients. Future studies of prediction and prognostic models should emphasise the quality of reporting, external model validation, generalisability to real clinical scenarios and enhanced reproducibility of results.
Collapse
Affiliation(s)
| | - Andy Welch
- Institute of Education in Healthcare and Medical Sciences, University of AberdeenAberdeenUK
| | | | - Mintu Nath
- Institute of Applied Health Sciences, University of AberdeenAberdeenUK
| |
Collapse
|
31
|
Pellegrino S, Fonti R, Hakkak Moghadam Torbati A, Bologna R, Morra R, Damiano V, Matano E, De Placido S, Del Vecchio S. Heterogeneity of Glycolytic Phenotype Determined by 18F-FDG PET/CT Using Coefficient of Variation in Patients with Advanced Non-Small Cell Lung Cancer. Diagnostics (Basel) 2023; 13:2448. [PMID: 37510192 PMCID: PMC10378511 DOI: 10.3390/diagnostics13142448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
We investigated the role of Coefficient of Variation (CoV), a first-order texture parameter derived from 18F-FDG PET/CT, in the prognosis of Non-Small Cell Lung Cancer (NSCLC) patients. Eighty-four patients with advanced NSCLC who underwent 18F-FDG PET/CT before therapy were retrospectively studied. SUVmax, SUVmean, CoV, total Metabolic Tumor Volume (MTVTOT) and whole-body Total Lesion Glycolysis (TLGWB) were determined by an automated contouring program (SUV threshold at 2.5). We analyzed 194 lesions: primary tumors (n = 84), regional (n = 48) and non-regional (n = 17) lymph nodes and metastases in liver (n = 9), bone (n = 23) and other sites (n = 13); average CoVs were 0.36 ± 0.13, 0.36 ± 0.14, 0.42 ± 0.18, 0.30 ± 0.14, 0.37 ± 0.17, 0.34 ± 0.13, respectively. No significant differences were found between the CoV values among the different lesion categories. Survival analysis included age, gender, histology, stage, MTVTOT, TLGWB and imaging parameters derived from primary tumors. At univariate analysis, CoV (p = 0.0184), MTVTOT (p = 0.0050), TLGWB (p = 0.0108) and stage (p = 0.0041) predicted Overall Survival (OS). At multivariate analysis, age, CoV, MTVTOT and stage were retained in the model (p = 0.0001). Patients with CoV > 0.38 had significantly better OS than those with CoV ≤ 0.38 (p = 0.0143). Patients with MTVTOT ≤ 89.5 mL had higher OS than those with MTVTOT > 89.5 mL (p = 0.0063). Combining CoV and MTVTOT, patients with CoV ≤ 0.38 and MTVTOT > 89.5 mL had the worst prognosis. CoV, by reflecting the heterogeneity of glycolytic phenotype, can predict clinical outcomes in NSCLC patients.
Collapse
Affiliation(s)
- Sara Pellegrino
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Rosa Fonti
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | | | - Roberto Bologna
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Rocco Morra
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131 Naples, Italy
| | - Vincenzo Damiano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131 Naples, Italy
| | - Elide Matano
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131 Naples, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", 80131 Naples, Italy
| | - Silvana Del Vecchio
- Department of Advanced Biomedical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
32
|
Liu W, Qiao X, Ge H, Zhang S, Sun X, Li J, Chen W, Gu W, Yuan S. Recurrence patterns are significantly associated with the 18F‑FDG PET/CT radiomic features of patients with locally advanced non‑small cell lung cancer treated with chemoradiotherapy. Oncol Lett 2023; 26:317. [PMID: 37332327 PMCID: PMC10272971 DOI: 10.3892/ol.2023.13903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023] Open
Abstract
A model for predicting the recurrence pattern of patients with locally advanced non-small cell lung cancer (LA-NSCLC) treated with chemoradiotherapy is of great importance for precision treatment. The present study analyzed whether the comprehensive quantitative values (CVs) of the fluorine-18(18F)-fluorodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) radiomic features and metastasis tumor volume (MTV) combined with clinical characteristics could predict the recurrence pattern of patients with LA-NSCLC treated with chemoradiotherapy. Patients with LA-NSCLC treated with chemoradiotherapy were divided into training and validation sets. The recurrence profile of each patient, including locoregional recurrence (LR), distant metastasis (DM) and both LR/DM were recorded. In the training set of patients, the primary tumor prior radiotherapy with 18F-FDG PET/CT and both primary tumors and lymph node metastasis were considered as the regions of interest (ROIs). The CVs of ROIs were calculated using principal component analysis. Additionally, MTVs were obtained from ROIs. The CVs, MTVs and the clinical characteristics of patients were subjected to aforementioned analysis. Furthermore, for the validation set of patients, the CVs and clinical characteristics of patients with LA-NSCLC were also subjected to logistic regression analysis and the area under the curve (AUC) values calculated. A total of 86 patients with LA-NSCLC were included in the analysis, including 59 and 27 patients in the training and validation sets of patients, respectively. The analysis revealed 22 and 12 cases with LR, 24 and 6 cases with DM and 13 and 9 cases with LR/DM in the training and validation sets of patients, respectively. Histological subtype, CV2-5 and CV3-4 were identified as independent variables in the logistic regression analysis (P<0.05). In addition, the AUC values for diagnosing LR, DM and LR/DM were 0.873, 0.711 and 0.826, and 0.675, 0.772 and 0.708 in the training and validation sets of patients, respectively. Overall, the results demonstrated that the spatial and metabolic heterogeneity quantitative values from the primary tumor combined with the histological subtype could predict the recurrence pattern of patients with LA-NSCLC treated with chemoradiotherapy.
Collapse
Affiliation(s)
- Wenju Liu
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong 250117, P.R. China
- Department of Radiation Oncology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Xu Qiao
- School of Control Science and Engineering, Shandong University, Jinan, Shandong 250117, P.R. China
| | - Hong Ge
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan 450000, P.R. China
| | - Sheng Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Xiaojiang Sun
- Department of Radiation Oncology, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Jiancheng Li
- Department of Radiation Oncology, Fujian Cancer Hospital, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian 350000, P.R. China
| | - Weilin Chen
- Department of Radiation Oncology, Zhangzhou Hospital Affiliated to Fujian Medical University, Zhangzhou, Fujian 363000, P.R. China
| | - Wendong Gu
- Department of Radiation Oncology, The Third Hospital Afiliated to Suzhou University, Changzhou, Jiangsu 213000, P.R. China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong 250117, P.R. China
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan 450000, P.R. China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| |
Collapse
|
33
|
Long ZC, Ding XC, Zhang XB, Sun PP, Hao FR, Li ZR, Hu M. The Efficacy of Pretreatment 18F-FDG PET-CT-Based Deep Learning Network Structure to Predict Survival in Nasopharyngeal Carcinoma. Clin Med Insights Oncol 2023; 17:11795549231171793. [PMID: 37251551 PMCID: PMC10214083 DOI: 10.1177/11795549231171793] [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: 11/28/2022] [Accepted: 04/10/2023] [Indexed: 05/31/2023] Open
Abstract
Background Previous studies have shown that the 5-year survival rates of patients with nasopharyngeal carcinoma (NPC) were still not ideal despite great improvement in NPC treatments. To achieve individualized treatment of NPC, we have been looking for novel models to predict the prognosis of patients with NPC. The objective of this study was to use a novel deep learning network structural model to predict the prognosis of patients with NPC and to compare it with the traditional PET-CT model combining metabolic parameters and clinical factors. Methods A total of 173 patients were admitted to 2 institutions between July 2014 and April 2020 for the retrospective study; each received a PET-CT scan before treatment. The least absolute shrinkage and selection operator (LASSO) was employed to select some features, including SUVpeak-P, T3, age, stage II, MTV-P, N1, stage III and pathological type, which were associated with overall survival (OS) of patients. We constructed 2 survival prediction models: an improved optimized adaptive multimodal task (a 3D Coordinate Attention Convolutional Autoencoder and an uncertainty-based jointly Optimizing Cox Model, CACA-UOCM for short) and a clinical model. The predictive power of these models was assessed using the Harrell Consistency Index (C index). Overall survival of patients with NPC was compared by Kaplan-Meier and Log-rank tests. Results The results showed that CACA-UOCM model could estimate OS (C index, 0.779 for training, 0.774 for validation, and 0.819 for testing) and divide patients into low and high mortality risk groups, which were significantly associated with OS (P < .001). However, the C-index of the model based only on clinical variables was only 0.42. Conclusions The deep learning network model based on 18F-FDG PET/CT can serve as a reliable and powerful predictive tool for NPC and provide therapeutic strategies for individual treatment.
Collapse
Affiliation(s)
- Zi-Chan Long
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xing-Chen Ding
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xian-Bin Zhang
- Department of General Surgery and Integrated Chinese and Western Medicine, Institute of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, China
| | - Peng-Peng Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fu-Rong Hao
- Department of Radiation Oncology, Weifang People's Hospital, Weifang, China
| | | | - Man Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
34
|
Chan KC, Perucho JAU, Subramaniam RM, Lee EYP. Utility of pre-treatment 18 F-fluorodeoxyglucose PET radiomic analysis in assessing nodal involvement in cervical cancer. Nucl Med Commun 2023; 44:375-380. [PMID: 36826394 DOI: 10.1097/mnm.0000000000001672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
OBJECTIVE Intratumor heterogeneity has prognostic value in cervical cancer, which can be depicted on 18 F-fluorodeoxyglucose ( 18 F-FDG) PET/computed tomography (PET/CT) and then quantitatively characterized by texture features. This study aimed to evaluate the discriminative performance and predictive ability of the texture features in determining lymph node involvement in cervical cancer. METHODS A total of 101 patients with newly diagnosed cervical cancer, who underwent pre-treatment whole-body 18 F-FDG PET/CT imaging were retrospectively recruited. Patients were categorized based on their nodal status. Thirty-five radiomic features together with the maximum standardized uptake value (SUVmax), metabolic tumor volume (MTV) and total lesion glycolysis (TLG) of the primary cervical tumors were extracted. Conventional indices were used to build logistic regression model and texture features were used to build random forest model. The performances for differentiating nodal status were assessed by receiver operating characteristic analysis. RESULTS Conventional PET indices were significantly higher in patients with nodal involvement compared to those without: SUVmax = 14.22 vs. 10.05; MTV = 57.02 vs. 28.73; TLG = 492.8 vs. 188.8 ( P < 0.05). Nineteen radiomic features describing regional heterogeneity were significantly different between nodal involvements. Area under the curves of the models with conventional indices and PET texture features for discriminating nodal status were 0.72 and 0.76, respectively. CONCLUSION PET-derived radiomic features had moderate performance in discriminating nodal involvement in cervical cancer; and they did not outperform model based on conventional indices.
Collapse
Affiliation(s)
- Kit Chi Chan
- Department of Radiotherapy, Hong Kong Sanatorium and Hospital
| | - Jose A U Perucho
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, Alabama
| | - Rathan M Subramaniam
- Department of Medicine, University of Otago, Dunedin, New Zealand
- Department of Radiology, Duke University, Durham, North Carolina, USA
| | - Elaine Y P Lee
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
35
|
Abenavoli EM, Barbetti M, Linguanti F, Mungai F, Nassi L, Puccini B, Romano I, Sordi B, Santi R, Passeri A, Sciagrà R, Talamonti C, Cistaro A, Vannucchi AM, Berti V. Characterization of Mediastinal Bulky Lymphomas with FDG-PET-Based Radiomics and Machine Learning Techniques. Cancers (Basel) 2023; 15:cancers15071931. [PMID: 37046592 PMCID: PMC10093023 DOI: 10.3390/cancers15071931] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/11/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND This study tested the diagnostic value of 18F-FDG PET/CT (FDG-PET) volumetric and texture parameters in the histological differentiation of mediastinal bulky disease due to classical Hodgkin lymphoma (cHL), primary mediastinal B-cell lymphoma (PMBCL) and grey zone lymphoma (GZL), using machine learning techniques. METHODS We reviewed 80 cHL, 29 PMBCL and 8 GZL adult patients with mediastinal bulky disease and histopathological diagnoses who underwent FDG-PET pre-treatment. Volumetric and radiomic parameters were measured using FDG-PET both for bulky lesions (BL) and for all lesions (AL) using LIFEx software (threshold SUV ≥ 2.5). Binary and multiclass classifications were performed with various machine learning techniques fed by a relevant subset of radiomic features. RESULTS The analysis showed significant differences between the lymphoma groups in terms of SUVmax, SUVmean, MTV, TLG and several textural features of both first- and second-order grey level. Among machine learning classifiers, the tree-based ensembles achieved the best performance both for binary and multiclass classifications in histological differentiation. CONCLUSIONS Our results support the value of metabolic heterogeneity as an imaging biomarker, and the use of radiomic features for early characterization of mediastinal bulky lymphoma.
Collapse
Affiliation(s)
- Elisabetta Maria Abenavoli
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| | - Matteo Barbetti
- Department of Information Engineering, University of Florence, 50134 Florence, Italy
- Istituto Nazionale di Fisica Nucleare (INFN), Florence Division, 50019 Sesto Fiorentino, Italy
| | - Flavia Linguanti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| | - Francesco Mungai
- Department of Radiology, Azienda Ospedaliero Universitaria Careggi, 50139 Florence, Italy
| | - Luca Nassi
- Hematology Department, Azienda Ospedaliero Universitaria Careggi, University of Florence, 50139 Florence, Italy
| | - Benedetta Puccini
- Hematology Department, Azienda Ospedaliero Universitaria Careggi, University of Florence, 50139 Florence, Italy
| | - Ilaria Romano
- Hematology Department, Azienda Ospedaliero Universitaria Careggi, University of Florence, 50139 Florence, Italy
| | - Benedetta Sordi
- Hematology Department, Azienda Ospedaliero Universitaria Careggi, University of Florence, 50139 Florence, Italy
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50139 Florence, Italy
| | - Raffaella Santi
- Pathology Section, Department of Health Sciences, University of Florence, 50139 Florence, Italy
| | - Alessandro Passeri
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| | - Roberto Sciagrà
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| | - Cinzia Talamonti
- Istituto Nazionale di Fisica Nucleare (INFN), Florence Division, 50019 Sesto Fiorentino, Italy
- Medical Physics Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| | - Angelina Cistaro
- Nuclear Medicine Department, Salus Alliance Medical, 16128 Genoa, Italy
- Pediatric Study Group for Italian Association of Nuclear Medicine (AIMN), 20159 Milan, Italy
| | - Alessandro Maria Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, 50139 Florence, Italy
| | - Valentina Berti
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, 50139 Florence, Italy
| |
Collapse
|
36
|
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
|
37
|
Heterogeneidad del tumor primario en la18F-FDG PET/TC pretratamiento para predecir el pronóstico en pacientes con cáncer de recto sometidos a cirugía tras terapia neoadyuvante. Rev Esp Med Nucl Imagen Mol 2023. [DOI: 10.1016/j.remn.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
38
|
Dong J, Gong Y, Liu Q, Wu Y, Fu F, Han H, Li X, Dong C, Wang M. Radiomics-based machine learning approach in differentiating fibro-adipose vascular anomaly from venous malformation. Pediatr Radiol 2023; 53:404-414. [PMID: 36271054 DOI: 10.1007/s00247-022-05520-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/05/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND As a complex vascular malformation, fibro-adipose vascular anomaly was first proposed in 2014. Its overlap with other vascular malformations regarding imaging and clinical features often leads to misdiagnosis and improper management. OBJECTIVE To construct a radiomics-based machine learning model to help radiologists differentiate fibro-adipose vascular anomaly from common venous malformations. MATERIALS AND METHODS We retrospectively analyzed 178 children, adolescents and young adults with vascular malformations (41 fibro-adipose vascular anomaly and 137 common vascular malformation cases) who underwent MRI before surgery between May 2012 to January 2021. We extracted radiomics features from T1-weighted images and fat-saturated (FS) T2-weighted images and further selected features through least absolute shrinkage and selection operator (LASSO) and Boruta methods. We established eight weighted logistic regression classification models based on various combinations of feature-selection strategies (LASSO or Boruta) and sequence types (single- or multi-sequence). Finally, we evaluated the performance of each model by the mean area under the receiver operating characteristics curve (ROC-AUC), sensitivity and specificity in 10 runs of repeated k-fold (k = 10) cross-validation. RESULTS Two multi-sequence models based on axial FS T2-W, coronal FS T2-W and axial T1-W images showed promising performance. The LASSO-based multi-sequence model achieved an AUC of 97%±3.8, a sensitivity of 94%±12.4 and a specificity of 89%±9.0. The Boruta-based multi-sequence model achieved an AUC of 97%±3.7, a sensitivity of 95%±10.5 and a specificity of 87%±9.0. CONCLUSION The radiomics-based machine learning model can provide a promising tool to help distinguish fibro-adipose vascular anomaly from common venous malformations.
Collapse
Affiliation(s)
- Jian Dong
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Yubin Gong
- Department of Hemangiomas and Vascular Malformations, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuyu Liu
- Department of Pathology, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Fangfang Fu
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Hui Han
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiaochen Li
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Changxian Dong
- Department of Hemangiomas and Vascular Malformations, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital & People's Hospital of Zhengzhou University, 7 Weiwu Road, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
39
|
Wongsa P, Nantasuk M, Singhnoi S, Pawano P, Jantarato A, Siripongsatian D, Lerdsirisuk P, Phonlakrai M. Assessing the variability and correlation between SUV and ADC parameters of head and neck cancers derived from simultaneous PET/MRI: A single-center study. J Appl Clin Med Phys 2023; 24:e13928. [PMID: 36763489 PMCID: PMC10161023 DOI: 10.1002/acm2.13928] [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: 08/06/2022] [Revised: 01/21/2023] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
OBJECTIVE Intratumoral heterogeneity is associated with poor outcomes in head and neck cancer (HNC) patients owing to chemoradiotherapy resistance. [18 F]-FDG positron emission tomography (PET) / Magnetic Resonance Imaging (MRI) provides spatial information about tumor mass, allowing intratumor heterogeneity assessment through histogram analysis. However, variability in quantitative PET/MRI parameter measurements could influence their reliability in assessing patient prognosis. Therefore, to use standardized uptake value (SUV) and apparent diffusion coefficient (ADC) parameters for assessing tumor response, this study aimed to measure SUV and ADC's variability and assess their relationship in HNC. METHODS First, ADC variability was measured in an in-house diffusion phantom and in five healthy volunteers. The SUV variability was only measured with the NEMA phantom using a clinical imaging protocol. Furthermore, simultaneous PET/MRI data of 11 HNC patients were retrospectively collected from the National Cyclotron and PET center in Chulabhorn Hospital. Tumor contours were manually drawn from PET images by an experienced nuclear medicine radiologist before tumor volume segmentation. Next, SUV and ADC's histogram were used to extract statistic variables of ADC and SUV: mean, median, min, max, skewness, kurtosis, and 5th , 10th , 25th , 50th , 75th , 90th , and 95th percentiles. Finally, the correlation between the statistic variables of ADC and SUV, as well as Metabolic Tumor volume and Total Lesion Glycolysis parameters was assessed using Pearson's correlation. RESULTS This pilot study showed that both parameters' maximum coefficient of variation was 13.9% and 9.8% in the phantom and in vivo, respectively. Furthermore, we found a strong and negative correlation between SUVmax and ADVmed (r = -0.75, P = 0.01). CONCLUSION The SUV and ADC obtained by simultaneous PET/MRI can be potentially used as an imaging biomarker for assessing intratumoral heterogeneity in patients with HNC. The low variability and relationship between SUV and ADC could allow multimodal prediction of tumor response in future studies.
Collapse
Affiliation(s)
- Paramest Wongsa
- School of Radiological Technology, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Sciences, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Mayurachat Nantasuk
- School of Radiological Technology, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Sciences, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Sinirun Singhnoi
- School of Radiological Technology, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Sciences, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Phattarasaya Pawano
- School of Radiological Technology, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Sciences, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Attapon Jantarato
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Pradith Lerdsirisuk
- National Cyclotron and PET Centre, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Monchai Phonlakrai
- School of Radiological Technology, Faculty of Health Science Technology, HRH Princess Chulabhorn College of Medical Sciences, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
40
|
Kihira S, Derakhshani A, Leung M, Mahmoudi K, Bauer A, Zhang H, Polson J, Arnold C, Tsankova NM, Hormigo A, Salehi B, Pham N, Ellingson BM, Cloughesy TF, Nael K. Multi-Parametric Radiomic Model to Predict 1p/19q Co-Deletion in Patients with IDH-1 Mutant Glioma: Added Value to the T2-FLAIR Mismatch Sign. Cancers (Basel) 2023; 15:cancers15041037. [PMID: 36831380 PMCID: PMC9954034 DOI: 10.3390/cancers15041037] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/28/2023] [Accepted: 01/28/2023] [Indexed: 02/10/2023] Open
Abstract
PURPOSE The T2-FLAIR mismatch sign has shown promise in determining IDH mutant 1p/19q non-co-deleted gliomas with a high specificity and modest sensitivity. To develop a multi-parametric radiomic model using MRI to predict 1p/19q co-deletion status in patients with newly diagnosed IDH1 mutant glioma and to perform a comparative analysis to T2-FLAIR mismatch sign+. METHODS In this retrospective study, patients with diagnosis of IDH1 mutant gliomas with known 1p/19q status who had preoperative MRI were included. T2-FLAIR mismatch was evaluated independently by two board-certified neuroradiologists. Texture features were extracted from glioma segmentation of FLAIR images. eXtremeGradient Boosting (XGboost) classifiers were used for model development. Leave-one-out-cross-validation (LOOCV) and external validation performances were reported for both the training and external validation sets. RESULTS A total of 103 patients were included for model development and 18 patients for external testing validation. The diagnostic performance (sensitivity/specificity/accuracy) in the determination of the 1p/19q co-deletion status was 59%/83%/67% (training) and 62.5%/70.0%/66.3% (testing) for the T2-FLAIR mismatch sign. This was significantly improved (p = 0.04) using the radiomics model to 77.9%/82.8%/80.3% (training) and 87.5%/89.9%/88.8% (testing), respectively. The addition of radiomics as a computer-assisted tool resulted in significant (p = 0.02) improvement in the performance of the neuroradiologist with 13 additional corrected cases in comparison to just using the T2-FLAIR mismatch sign. CONCLUSION The proposed radiomic model provides much needed sensitivity to the highly specific T2-FLAIR mismatch sign in the determination of the 1p/19q non-co-deletion status and improves the overall diagnostic performance of neuroradiologists when used as an assistive tool.
Collapse
Affiliation(s)
- Shingo Kihira
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Ahrya Derakhshani
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Michael Leung
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Keon Mahmoudi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Adam Bauer
- Department of Radiology, Kaiser Permanente Fontana Medical Center, Fontana, CA 92335, USA
| | - Haoyue Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jennifer Polson
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Corey Arnold
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Nadejda M. Tsankova
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adilia Hormigo
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Banafsheh Salehi
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Nancy Pham
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
| | - Benjamin M. Ellingson
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Timothy F. Cloughesy
- UCLA Brain Tumor Imaging Laboratory, Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Neurology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kambiz Nael
- Department of Radiological Sciences, David Geffen School of Medicine at University of California Los Angeles, 757 Westwood Plaza, Suite 1621, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-310-267-5932
| |
Collapse
|
41
|
Park SB, Kim KU, Park YW, Hwang JH, Lim CH. Application of 18 F-fluorodeoxyglucose PET/CT radiomic features and machine learning to predict early recurrence of non-small cell lung cancer after curative-intent therapy. Nucl Med Commun 2023; 44:161-168. [PMID: 36458424 DOI: 10.1097/mnm.0000000000001646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
OBJECTIVE To predict the recurrence of non-small cell lung cancer (NSCLC) within 2 years after curative-intent treatment using a machine-learning approach with PET/CT-based radiomics. PATIENTS AND METHODS A total of 77 NSCLC patients who underwent pretreatment 18 F-fluorodeoxyglucose PET/CT were retrospectively analyzed. Five clinical features (age, sex, tumor stage, tumor histology, and smoking status) and 48 radiomic features extracted from primary tumors on PET were used for binary classifications. These were ranked, and a subset of useful features was selected based on Gini coefficient scores in terms of associations with relapsed status. Areas under the receiver operating characteristics curves (AUC) were yielded by six machine-learning algorithms (support vector machine, random forest, neural network, naive Bayes, logistic regression, and gradient boosting). Model performances were compared and validated via random sampling. RESULTS A PET/CT-based radiomic model was developed and validated for predicting the recurrence of NSCLC during the first 2 years after curation. The most important features were SD and variance of standardized uptake value, followed by low-intensity short-zone emphasis and high-intensity zone emphasis. The naive Bayes model with the 15 best-ranked features displayed the best performance (AUC: 0.816). Prediction models using the five best PET-derived features outperformed those using five clinical variables. CONCLUSION The machine learning model using PET-derived radiomic features showed good performance for predicting the recurrence of NSCLC during the first 2 years after a curative intent therapy. PET/CT-based radiomic features may help clinicians improve the risk stratification of relapsed NSCLC.
Collapse
Affiliation(s)
| | - Ki-Up Kim
- Department of Allergy and Respiratory Medicine
| | | | - Jung Hwa Hwang
- Department of Radiology, Soonchunhyang University Hospital, Seoul, Republic of Korea
| | | |
Collapse
|
42
|
Hu S, Kang Y, Xie Y, Yang T, Yang Y, Jiao J, Zou Q, Zhang H, Zhang Y. 18F-FDG PET/CT-based radiomics nomogram for preoperative prediction of macrotrabecular-massive hepatocellular carcinoma: a two-center study. Abdom Radiol (NY) 2023; 48:532-542. [PMID: 36370179 DOI: 10.1007/s00261-022-03722-y] [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: 08/25/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/13/2022]
Abstract
PURPOSE To explore the potential of β-2-[18F] fluoro-2-deoxy-D-glucose positron emission tomography/computed tomography (18F-FDG PET/CT) in the evaluation of macrotrabecular-massive (MTM) hepatocellular carcinoma (HCC) and to apply radiomics approach to build a radiomics nomogram for predicting MTM-HCC. METHODS This study included 140 (training cohort:101; validation cohort:39) HCC patients who underwent preoperative 18F-FDG PET/CT at two institutions. The clinical features and tumor FDG metabolism measured by the tumor-to-liver ratio (TLR) via 18F-FDG PET/CT were retrospectively collected. Radiomics features were extracted from 18F-FDG PET/CT images and a radiomics score (Rad-score) was calculated. A radiomics nomogram was then constructed by combining Rad-score and independent clinical features and was assessed with a calibration curve. The performance of the radiomics nomogram, Rad-score and TLR was evaluated by receiver operating characteristic (ROC) curves and decision curve analysis (DCA). RESULTS A total of six top weighted radiomics features were selected from PET/CT images by the least absolute shrinkage and selection operator (LASSO) regression algorithm and were used to construct a Rad-score. Multivariate analysis identified Rad-score (OR = 2.183, P = 0.004), age ≤ 50 years (OR = 3.136, P = 0.036), AST > 40U/L (OR = 0.270, P = 0.017) and TLR (OR = 1.641, P = 0.049) as independent predictors of MTM-HCC. The radiomics nomogram had a higher area under the curves (AUCs) than the Rad-score and TLR for predicting MTM-HCC in both training (0.849 [95% CI 0.774-0.924] vs. 0.764 [95% CI 0.669-0.843], 0.763 [95% CI 0.668-0.842]) and validation (0.749 [95% CI 0.584-0.873] vs. 0.690 [95% CI 0.522-0.828], 0.541 [95% CI 0.374-0.701]) cohorts. DCA showed the radiomics nomogram to be more clinically useful than Rad-score and TLR. CONCLUSIONS Tumor FDG metabolism is significantly associated with MTM-HCC. A 18F-FDG PET/CT-based radiomics nomogram may be useful for preoperatively predicting the MTM subtype in primary HCC patients, contributing to pretreatment decision-making.
Collapse
Affiliation(s)
- Siqi Hu
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yinqian Kang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, No. 651 Dongfeng East Road, Yuexiu District, Guangzhou, 510060, China
| | - Yujie Xie
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Ting Yang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Yuan Yang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Ju Jiao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Qiong Zou
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China
| | - Hong Zhang
- Department of Nuclear Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 33 Yingfeng Road, Haizhu District, Guangzhou, 510289, China.
| | - Yong Zhang
- Department of Nuclear Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600 Tianhe Road, Tianhe District, Guangzhou, 510630, China.
| |
Collapse
|
43
|
Ju HM, Kim BC, Lim I, Byun BH, Woo SK. Estimation of an Image Biomarker for Distant Recurrence Prediction in NSCLC Using Proliferation-Related Genes. Int J Mol Sci 2023; 24:ijms24032794. [PMID: 36769108 PMCID: PMC9917349 DOI: 10.3390/ijms24032794] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/22/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
This study aimed to identify a distant-recurrence image biomarker in NSCLC by investigating correlations between heterogeneity functional gene expression and fluorine-18-2-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) image features of NSCLC patients. RNA-sequencing data and 18F-FDG PET images of 53 patients with NSCLC (19 with distant recurrence and 34 without recurrence) from The Cancer Imaging Archive and The Cancer Genome Atlas Program databases were used in a combined analysis. Weighted correlation network analysis was performed to identify gene groups related to distant recurrence. Genes were selected for functions related to distant recurrence. In total, 47 image features were extracted from PET images as radiomics. The relationship between gene expression and image features was estimated using a hypergeometric distribution test with the Pearson correlation method. The distant recurrence prediction model was validated by a random forest (RF) algorithm using image texture features and related gene expression. In total, 37 gene modules were identified by gene-expression pattern with weighted gene co-expression network analysis. The gene modules with the highest significance were selected (p-value < 0.05). Nine genes with high protein-protein interaction and area under the curve (AUC) were identified as hub genes involved in the proliferation function, which plays an important role in distant recurrence of cancer. Four image features (GLRLM_SRHGE, GLRLM_HGRE, SUVmean, and GLZLM_GLNU) and six genes were identified to be correlated (p-value < 0.1). AUCs (accuracy: 0.59, AUC: 0.729) from the 47 image texture features and AUCs (accuracy: 0.767, AUC: 0.808) from hub genes were calculated using the RF algorithm. AUCs (accuracy: 0.783, AUC: 0.912) from the four image texture features and six correlated genes and AUCs (accuracy: 0.738, AUC: 0.779) from only the four image texture features were calculated using the RF algorithm. The four image texture features validated by heterogeneity group gene expression were found to be related to cancer heterogeneity. The identification of these image texture features demonstrated that advanced prediction of NSCLC distant recurrence is possible using the image biomarker.
Collapse
Affiliation(s)
- Hye Min Ju
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Byung-Chul Kim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Ilhan Lim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Byung Hyun Byun
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
| | - Sang-Keun Woo
- Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon 34113, Republic of Korea
- Division of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 07812, Republic of Korea
- Correspondence: ; Tel.: +82-2-970-1659
| |
Collapse
|
44
|
Gülbahar Ateş S, Bilir Dilek G, Uçmak G. Primary tumor heterogeneity on pretreatment 18F-FDG PET/CT to predict outcome in patients with rectal cancer who underwent surgery after neoadjuvant therapy. Rev Esp Med Nucl Imagen Mol 2023:S2253-8089(23)00001-0. [PMID: 36690032 DOI: 10.1016/j.remnie.2023.01.001] [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: 10/17/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/21/2023]
Abstract
PURPOSE This retrospective study aimed to investigate the value of texture features of primary tumors in pretreatment 18F-FDG PET/CT in the prediction of response to treatment, progression, and overall survival in patients with rectal cancer who underwent surgery after neoadjuvant therapy(NAT). METHODS Patients with rectal cancer who had pretreatment 18F-FDG PET/CT, and underwent surgery after NAT were included in this study. Clinicopathologic features, date of last follow-up, progression, and death were recorded. Textural and conventional PET parameters(maximum standardized uptake value-SUVmax, metabolic tumor volume-MTV, total lesion glycolysis-TLG) were obtained from PET/CT images using LifeX program. Parameters were grouped using Youden index in ROC analysis. Factors predicting the pathological response to treatment, progression, and overall survival were determined using logistic regression and Cox regression analyses. RESULTS Forty-four patients (26(59%) male, 18(41%) female; 60.1±11.4 years) with rectal cancer were included in this study. The numbers of patients with responders and non-responders to NAT were 15(34.9%) and 28(65.1%), respectively. One patient' pathology report did not contain the response status to NAT. The median of follow-up duration was 29.9 months. 9(20.5%) showed disease progression, and 8(18.2%) died during the follow-up period. Difference entropyGLCM and correlationGLCM parameters were found as independent predictors for response to NAT. The positivity of surgical margin, intensity interquartile rangeCONV and AUC-CSHDISC texture parameters were independent predictors of progression, while normalized inverse differenceGLCM and LZLGEGLZLM parameters were independent predictors of mortality. CONCLUSION The texture parameters obtained from pretreatment 18F-FDG PET/CT have presented a more robust predictive value than conventional parameters in patients with rectal cancer who underwent surgery after NAT.
Collapse
Affiliation(s)
- Seda Gülbahar Ateş
- Department of Nuclear Medicine, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, University of Health Sciences, Ankara, Turkey.
| | - Gülay Bilir Dilek
- Department of Pathology, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| | - Gülin Uçmak
- Department of Nuclear Medicine, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, University of Health Sciences, Ankara, Turkey
| |
Collapse
|
45
|
Xu L, Yang X, Xiang W, Hu P, Zhang X, Li Z, Li Y, Liu Y, Dai Y, Luo Y, Qiu H. Development and validation of a contrast-enhanced CT-based radiomics nomogram for preoperative diagnosis in neuroendocrine carcinoma of digestive system. Front Endocrinol (Lausanne) 2023; 14:1155307. [PMID: 37124722 PMCID: PMC10130364 DOI: 10.3389/fendo.2023.1155307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/28/2023] [Indexed: 05/02/2023] Open
Abstract
Objectives To develop and validate a contrast-enhanced CT-based radiomics nomogram for the diagnosis of neuroendocrine carcinoma of the digestive system. Methods The clinical data and contrast-enhanced CT images of 60 patients with pathologically confirmed neuroendocrine carcinoma of the digestive system and 60 patients with non-neuroendocrine carcinoma of the digestive system were retrospectively collected from August 2015 to December 2021 at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, and randomly divided into a training cohort (n=84) and a validation cohort (n=36). Clinical characteristics were analyzed by logistic regression and a clinical diagnosis model was developed. Radiomics signature were established by extracting radiomic features from contrast-enhanced CT images. Based on the radiomic signature and clinical characteristics, radiomic nomogram was developed. ROC curves and Delong's test were used to evaluate the diagnostic efficacy of the three models, calibration curves and application decision curves were used to analyze the accuracy and clinical application value of nomogram. Results Logistic regression results showed that TNM stage (stage IV) (OR 6.8, 95% CI 1.320-43.164, p=0. 028) was an independent factor affecting the diagnosis for NECs of the digestive system, and a clinical model was constructed based on TNM stage (stage IV). The AUCs of the clinical model, radiomics signature, and radiomics nomogram for the diagnosis of NECs of the digestive system in the training, validation cohorts and pooled patients were 0.643, 0.893, 0.913; 0.722, 0.867, 0.932 and 0.667, 0.887, 0.917 respectively. The AUCs of radiomics signature and radiomics nomogram were higher than clinical model, with statistically significant difference (Z=4.46, 6.85, both p < 0.001); the AUC difference between radiomics signature and radiomics nomogram was not statistically significant (Z=1.63, p = 0.104). The results of the calibration curve showed favorable agreement between the predicted values of the nomogram and the pathological results, and the decision curve analysis indicated that the nomogram had favorable application in clinical practice. Conclusions The nomogram constructed based on contrast-enhanced CT radiomics and clinical characteristics was able to effectively diagnose neuroendocrine carcinoma of the digestive system.
Collapse
Affiliation(s)
- Liang Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxuan Xiang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengbo Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuyuan Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiming Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongqing Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhong Dai
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Luo
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hong Qiu, ; Yan Luo,
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hong Qiu, ; Yan Luo,
| |
Collapse
|
46
|
Jiao W, Song S, Han H, Wang W, Zhang Q. Artificially intelligent differential diagnosis of enlarged lymph nodes with random vector functional link network plus. Med Eng Phys 2023; 111:103939. [PMID: 36792248 DOI: 10.1016/j.medengphy.2022.103939] [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: 06/23/2022] [Revised: 11/10/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
Differential diagnosis of enlarged lymph nodes (ELNs) is essential for the treatment of related patients. Though multi-modal ultrasound including B-mode, Doppler ultrasound, elastography and contrast-enhanced ultrasound (CEUS) can enhance diagnostic performance for ELNs, the scenario of having only single or dual modal data is often encountered. In this study, an artificially intelligent diagnosis model based on the learning using privileged information was proposed to aid in differential diagnosis of ELNs in the case of single or dual modal images. In our model, B-mode, or combined with another modality, was used as the standard information (SI) and other modalities were used as the privileged information (PI). The model was constructed through the combination of the SI and PI in the training stage. By learning from the training samples, a random vector functional link network with privileged information (RVFL+) was obtained, which was used to classify the testing samples of solely the SI. Results showed that the accuracy, precision and Youden's index of the RVFL+ model, using B-mode with elastography as the SI and CEUS as the PI, reached 78.4%, 92.4% and 54.9%, increased by 14.0%, 8.4% and 24.5% compared with the model using B-mode as the SI without the PI. The method based on the LUPI can improve the diagnostic performance for ELNs.
Collapse
Affiliation(s)
- Weiwei Jiao
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Shuang Song
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Hong Han
- Department of Ultrasound, Zhongshan Hospital Fudan University, 200032, Shanghai, China; Shanghai Institute of Medical Imaging, 200032, Shanghai, China.
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital Fudan University, 200032, Shanghai, China; Shanghai Institute of Medical Imaging, 200032, Shanghai, China.
| | - Qi Zhang
- The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China; School of Communication and Information Engineering, Shanghai University, Shanghai, China.
| |
Collapse
|
47
|
Hu Q, Li K, Yang C, Wang Y, Huang R, Gu M, Xiao Y, Huang Y, Chen L. The role of artificial intelligence based on PET/CT radiomics in NSCLC: Disease management, opportunities, and challenges. Front Oncol 2023; 13:1133164. [PMID: 36959810 PMCID: PMC10028142 DOI: 10.3389/fonc.2023.1133164] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Objectives Lung cancer has been widely characterized through radiomics and artificial intelligence (AI). This review aims to summarize the published studies of AI based on positron emission tomography/computed tomography (PET/CT) radiomics in non-small-cell lung cancer (NSCLC). Materials and methods A comprehensive search of literature published between 2012 and 2022 was conducted on the PubMed database. There were no language or publication status restrictions on the search. About 127 articles in the search results were screened and gradually excluded according to the exclusion criteria. Finally, this review included 39 articles for analysis. Results Classification is conducted according to purposes and several studies were identified at each stage of disease:1) Cancer detection (n=8), 2) histology and stage of cancer (n=11), 3) metastases (n=6), 4) genotype (n=6), 5) treatment outcome and survival (n=8). There is a wide range of heterogeneity among studies due to differences in patient sources, evaluation criteria and workflow of radiomics. On the whole, most models show diagnostic performance comparable to or even better than experts, and the common problems are repeatability and clinical transformability. Conclusion AI-based PET/CT Radiomics play potential roles in NSCLC clinical management. However, there is still a long way to go before being translated into clinical application. Large-scale, multi-center, prospective research is the direction of future efforts, while we need to face the risk of repeatability of radiomics features and the limitation of access to large databases.
Collapse
Affiliation(s)
- Qiuyuan Hu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Conghui Yang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yue Wang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Rong Huang
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Mingqiu Gu
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yuqiang Xiao
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| | - Long Chen
- Department of positron emission tomography/computed tomography (PET/CT) Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, Yunnan, China
- *Correspondence: Long Chen, ; Yunchao Huang,
| |
Collapse
|
48
|
Zhang Y, Zhang Y, Peng L, Zhang L. Research Progress on the Predicting Factors and Coping Strategies for Postoperative Recurrence of Esophageal Cancer. Cells 2022; 12:cells12010114. [PMID: 36611908 PMCID: PMC9818463 DOI: 10.3390/cells12010114] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 12/01/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
Esophageal cancer is one of the malignant tumors with poor prognosis in China. Currently, the treatment of esophageal cancer is still based on surgery, especially in early and mid-stage patients, to achieve the goal of radical cure. However, esophageal cancer is a kind of tumor with a high risk of recurrence and metastasis, and locoregional recurrence and distant metastasis are the leading causes of death after surgery. Although multimodal comprehensive treatment has advanced in recent years, the prediction, prevention and treatment of postoperative recurrence and metastasis of esophageal cancer are still unsatisfactory. How to reduce recurrence and metastasis in patients after surgery remains an urgent problem to be solved. Given the clinical demand for early detection of postoperative recurrence of esophageal cancer, clinical and basic research aiming to meet this demand has been a hot topic, and progress has been observed in recent years. Therefore, this article reviews the research progress on the factors that influence and predict postoperative recurrence of esophageal cancer, hoping to provide new research directions and treatment strategies for clinical practice.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Yuxin Zhang
- Department of Pediatric Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Lin Peng
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, China
- Correspondence:
| |
Collapse
|
49
|
Assadi M, Manafi-Farid R, Jafari E, Keshavarz A, Divband G, Moradi MM, Adinehpour Z, Samimi R, Dadgar H, Jokar N, Mayer B, Prasad V. Predictive and prognostic potential of pretreatment 68Ga-PSMA PET tumor heterogeneity index in patients with metastatic castration-resistant prostate cancer treated with 177Lu-PSMA. Front Oncol 2022; 12:1066926. [PMID: 36568244 PMCID: PMC9773988 DOI: 10.3389/fonc.2022.1066926] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION This study was conducted to evaluate the predictive values of volumetric parameters and radiomic features (RFs) extracted from pretreatment 68Ga-PSMA PET and baseline clinical parameters in response to 177Lu-PSMA therapy. MATERIALS AND METHODS In this retrospective multicenter study, mCRPC patients undergoing 177Lu-PSMA therapy were enrolled. According to the outcome of therapy, the patients were classified into two groups including positive biochemical response (BCR) (≥ 50% reduction in the serum PSA value) and negative BCR (< 50%). Sixty-five RFs, eight volumetric parameters, and also seventeen clinical parameters were evaluated for the prediction of BCR. In addition, the impact of such parameters on overall survival (OS) was evaluated. RESULTS 33 prostate cancer patients with a median age of 69 years (range: 49-89) were enrolled. BCR was observed in 22 cases (66%), and 16 cases (48.5%) died during the follow-up time. The results of Spearman correlation test indicated a significant relationship between BCR and treatment cycle, administered dose, HISTO energy, GLCM entropy, and GLZLM LZLGE (p<0.05). In addition, according to the Mann-Whitney U test, age, cycle, dose, GLCM entropy, and GLZLM LZLGE were significantly different between BCR and non BCR patients (p<0.05). According to the ROC curve analysis for feature selection for prediction of BCR, GLCM entropy, age, treatment cycle, and administered dose showed acceptable results (p<0.05). According to SVM for assessing the best model for prediction of response to therapy, GLCM entropy alone showed the highest predictive performance in treatment planning. For the entire cohort, the Kaplan-Meier test revealed a median OS of 21 months (95% CI: 12.12-29.88). The median OS was estimated at 26 months (95% CI: 17.43-34.56) for BCR patients and 13 months (95% CI: 9.18-16.81) for non BCR patients. Among all variables included in the Kaplan Meier, the only response to therapy was statistically significant (p=0.01). CONCLUSION This exploratory study showed that the heterogeneity parameter of pretreatment 68Ga-PSMA PET images might be a potential predictive value for response to 177Lu-PSMA therapy in mCRPC; however, further prospective studies need to be carried out to verify these findings.
Collapse
Affiliation(s)
- Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Nuclear Medicine, Molecular Imaging, and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Esmail Jafari
- The Persian Gulf Nuclear Medicine Research Center, Department of Nuclear Medicine, Molecular Imaging, and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Ahmad Keshavarz
- IoT and Signal Processing Research Group, ICT Research Institute, Faculty of Intelligent Systems Engineering and Data Science, Persian Gulf University, Bushehr, Iran
| | | | - Mohammad Mobin Moradi
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Rezvan Samimi
- Department of Medical Radiation Engineering, Shahid Beheshti University, Tehran, Iran
| | - Habibollah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad, Iran
| | - Narges Jokar
- The Persian Gulf Nuclear Medicine Research Center, Department of Nuclear Medicine, Molecular Imaging, and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Vikas Prasad
- Department of Nuclear Medicine, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
50
|
Imaging-based representation and stratification of intra-tumor heterogeneity via tree-edit distance. Sci Rep 2022; 12:19607. [PMID: 36380083 PMCID: PMC9666477 DOI: 10.1038/s41598-022-23752-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 11/04/2022] [Indexed: 11/17/2022] Open
Abstract
Personalized medicine is the future of medical practice. In oncology, tumor heterogeneity assessment represents a pivotal step for effective treatment planning and prognosis prediction. Despite new procedures for DNA sequencing and analysis, non-invasive methods for tumor characterization are needed to impact on daily routine. On purpose, imaging texture analysis is rapidly scaling, holding the promise to surrogate histopathological assessment of tumor lesions. In this work, we propose a tree-based representation strategy for describing intra-tumor heterogeneity of patients affected by metastatic cancer. We leverage radiomics information extracted from PET/CT imaging and we provide an exhaustive and easily readable summary of the disease spreading. We exploit this novel patient representation to perform cancer subtyping according to hierarchical clustering technique. To this purpose, a new heterogeneity-based distance between trees is defined and applied to a case study of prostate cancer. Clusters interpretation is explored in terms of concordance with severity status, tumor burden and biological characteristics. Results are promising, as the proposed method outperforms current literature approaches. Ultimately, the proposed method draws a general analysis framework that would allow to extract knowledge from daily acquired imaging data of patients and provide insights for effective treatment planning.
Collapse
|