1
|
Deng H, Zhao L, Ge H, Gao Y, Fu Y, Lin Y, Masoodi M, Losmanova T, Medová M, Ott J, Su M, Wang W, Peng RW, Dorn P, Marti TM. Ubiquinol-mediated suppression of mitochondria-associated ferroptosis is a targetable function of lactate dehydrogenase B in cancer. Nat Commun 2025; 16:2597. [PMID: 40090955 PMCID: PMC11911438 DOI: 10.1038/s41467-025-57906-3] [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: 07/31/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Lactate dehydrogenase B (LDHB) fuels oxidative cancer cell metabolism by converting lactate to pyruvate. This study uncovers LDHB's role in countering mitochondria-associated ferroptosis independently of lactate's function as a carbon source. LDHB silencing alters mitochondrial morphology, causes lipid peroxidation, and reduces cancer cell viability, which is potentiated by the ferroptosis inducer RSL3. Unlike LDHA, LDHB acts in parallel with glutathione peroxidase 4 (GPX4) and dihydroorotate dehydrogenase (DHODH) to suppress mitochondria-associated ferroptosis by decreasing the ubiquinone (coenzyme Q, CoQ) to ubiquinol (CoQH2) ratio. Indeed, supplementation with mitoCoQH2 (mitochondria-targeted analogue of CoQH2) suppresses mitochondrial lipid peroxidation and cell death after combined LDHB silencing and RSL3 treatment, consistent with the presence of LDHB in the cell fraction containing the mitochondrial inner membrane. Addressing the underlying molecular mechanism, an in vitro NADH consumption assay with purified human LDHB reveals that LDHB catalyzes the transfer of reducing equivalents from NADH to CoQ and that the efficiency of this reaction increases by the addition of lactate. Finally, radiation therapy induces mitochondrial lipid peroxidation and reduces tumor growth, which is further enhanced when combined with LDHB silencing. Thus, LDHB-mediated lactate oxidation drives the CoQ-dependent suppression of mitochondria-associated ferroptosis, a promising target for combination therapies.
Collapse
Affiliation(s)
- Haibin Deng
- 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for esophageal carcinoma, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Liang Zhao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Huixiang Ge
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Yan Fu
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Yantang Lin
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Mojgan Masoodi
- Institute of Clinical Chemistry, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Tereza Losmanova
- Institute of Tissue Medicine and Pathology, ITMP, University of Bern, Bern, Switzerland
| | - Michaela Medová
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for esophageal carcinoma, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Julien Ott
- Department of Radiation Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Min Su
- 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for esophageal carcinoma, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Wenxiang Wang
- 2nd Department of Thoracic Surgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
- Hunan Clinical Medical Research Center of Accurate Diagnosis and Treatment for esophageal carcinoma, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
2
|
Wu J, Huang ZN, Zhang XQ, Hou SS, Wang JB, Chen QY, Li P, Xie JW, Huang CM, Lin JX, Zheng CH. Development of a modified nutritional index model based on nutritional status and sarcopenia to predict long-term survival and chemotherapy benefits in elderly patients with advanced gastric cancer. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109503. [PMID: 39642588 DOI: 10.1016/j.ejso.2024.109503] [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: 07/13/2024] [Revised: 11/04/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Elderly patients with advanced gastric cancer have poor prognoses. This study aims to develop a prediction model for long-term survival after radical surgery and to identify patients who may benefit from chemotherapy. METHODS Data from 555 elderly patients with advanced gastric cancer admitted to two medical centers from 2009 to 2018 were retrospectively analyzed. Sarcopenia was combined with the Controlling Nutritional Status (CONUT) score to create a modified nutritional index (mCONUT). Cox regression analyses were used to develop a novel nomogram prediction model (mCNS) that combined mCONUT, pN, and tumor size, and its performance was further verified both internally and externally. RESULTS Multivariate Cox analysis revealed that tumor size, pN, and mCONUT were independent prognostic risk factors for overall survival (OS). The mCNS model showed good fit and high predictive value (AUC: training set 0.711; validation set 0.707), outperforming the pTNM model (p < 0.05). To further investigate the association between the model and adjuvant chemotherapy, we categorized the model into two risk groups: a high-risk group and a low-risk group. Further analysis revealed that, in the low-risk group, the OS and recurrence-free survival(RFS) for patients receiving adjuvant chemotherapy was significantly better than that of those who did not receive chemotherapy (p = 0.047,p = 0.019). In the high-risk group, this result was not observed (p = 0.120, p = 0.053). CONCLUSION The mCNS model has high predictive value in predicting long-term survival of elderly patients with advanced gastric cancer. Patients with mCNS-L were able to benefit from chemotherapy after laparoscopic radical gastrectomy.
Collapse
Affiliation(s)
- Ju Wu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ze-Ning Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xing-Qi Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shuang-Shuang Hou
- Department of General Surgery, Affiliated Zhongshan Hospital of Dalian University, Dalian, China; Department of Surgery, FuYang Normal University Second Affiliated Hospital, Fuyang, China
| | - Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
3
|
Seo D, Ito R, Ishikawa K, Miura T, Yamamoto Y, Onodera Y, Nishioka S, Ito YM, Fuyama K, Maeda T. 3D scanner's potential as a novel tool for lymphedema measurement in mouse hindlimb models. Sci Rep 2025; 15:3747. [PMID: 39885185 PMCID: PMC11782579 DOI: 10.1038/s41598-025-85637-4] [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: 04/20/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
Lymphedema is characterized by persistent swelling due to impaired lymphatic function and presents significant challenges in both research and clinical settings. Traditional contact-based measurement techniques such as paw thickness and circumferential measurements using calipers or silk thread are useful but limited by observer variability and measurement accuracy. Non-contact methods, including various imaging techniques, offer improvements but often at higher cost and complexity. In this study, we address the need for a more reliable, cost-effective, and non-invasive method for assessing lymphedema in mouse models. Here we show that 3D scanning technology can enhance the measurement of lymphedema in a mouse hindlimb model. Our results indicate that 3D scanners provide more consistent measurements with lower variability compared with conventional methods and without the need for direct contact, which could potentially alter the measurement outcomes. The findings of this study suggest that 3D scanning could replace traditional methods, offering a more standardized and less subjective tool for lymphedema research in the near future. This technology would not only improve upon conventional methods but also extend the capabilities for detailed anatomical analyses in small animal models, which could have implications for other areas of biomedical research.
Collapse
Affiliation(s)
- Dongkyung Seo
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Riri Ito
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Soichiro Nishioka
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoichi M Ito
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Kanako Fuyama
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
4
|
Nogueira SA, Luz FAB, Camargo TFO, Oliveira JCS, Campos Neto GC, Carvalhaes FBF, Reis MRC, Santos PV, Mendes GS, Loureiro RM, Tornieri D, Pacheco VMG, Coimbra AP, Calixto WP. Artificial intelligence applied in identifying left ventricular walls in myocardial perfusion scintigraphy images: Pilot study. PLoS One 2025; 20:e0312257. [PMID: 39823407 PMCID: PMC11741626 DOI: 10.1371/journal.pone.0312257] [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: 04/18/2024] [Accepted: 10/03/2024] [Indexed: 01/19/2025] Open
Abstract
This paper proposes the use of artificial intelligence techniques, specifically the nnU-Net convolutional neural network, to improve the identification of left ventricular walls in images of myocardial perfusion scintigraphy, with the objective of improving the diagnosis and treatment of coronary artery disease. The methodology included data collection in a clinical environment, followed by data preparation and analysis using the 3D Slicer Platform for manual segmentation, and subsequently, the application of artificial intelligence models for automated segmentation, focusing on the efficiency of identifying the walls of the left ventricular. A total of 83 clinical routine exams were collected, each exam containing 50 slices, which is 4,150 images. The results demonstrate the efficiency of the proposed artificial intelligence model, with a Dice coefficient of 87% and an average Intersection over Union of 0.8, reflecting high agreement with the manual segmentations produced by experts and surpassing traditional interpretation methods. The internal and external validation of the model corroborates its future applicability in real clinical scenarios, offering a new perspective in the analysis of myocardial perfusion scintigraphy images. The integration of artificial intelligence into the process of analyzing myocardial perfusion scintigraphy images represents a significant advancement in diagnostic accuracy, promoting substantial improvements in the interpretation of medical images, and establishing a foundation for future research and clinical applications, such as artifact correction.
Collapse
Affiliation(s)
- Solange Amorim Nogueira
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | - Fernanda Ambrogi B. Luz
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | - Thiago Fellipe O. Camargo
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | | | | | | | - Marcio Rodrigues C. Reis
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | - Paulo Victor Santos
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | | | | | | | - Viviane M. Gomes Pacheco
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| | | | - Wesley Pacheco Calixto
- Electrical, Mechanical & Computer Engineering School, Federal University of Goias, Goiania, Brazil
- Systems and Robotics Institute, Coimbra University, Coimbra, Portugal
- Technology Research and Development Center (GCITE), Federal Institute of Goias, Goiania, Goias, Brazil
| |
Collapse
|
5
|
Liu C, Zhao J, Zhang H, Ni X. Diagnostic Value of 18F-FDG PET/CT Radiomics in Lymphoma: A Systematic Review and Meta-Analysis. Technol Cancer Res Treat 2025; 24:15330338251342860. [PMID: 40397102 PMCID: PMC12099142 DOI: 10.1177/15330338251342860] [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: 02/11/2025] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/22/2025] Open
Abstract
IntroductionVarious machine learning models and features have been proposed for lymphoma diagnosis using 18F-fluorodeoxyglucose (18F-FDG) PET/CT radiomics. This research aimed to systematically evaluate the diagnostic value of 18F-FDG PET/CT radiomics in lymphoma by conducting a meta-analysis.MethodsData from published studies regarding the diagnosis of lymphoma using 18F-FDG PET/CT radiomics, from January 2010 to July 2024, were gathered from PubMed, Web of Science, and the Cochrane Library. Following their separate searches and screenings of the literature, two researchers extracted data and assessed the caliber of all the included studies. The quality assessment involved the Quality Assessment of Diagnostic Accuracy Studies 2 (QUADAS-2), the Radiomics Quality Score (RQS), and the METhodological RadiomICs Score (METRICS). The meta-analysis was conducted by using RevMan 5.4.1, R 4.4.0, and Stata 17.0 software. Six meta-regressions were conducted on study performance, considering sample size, image modality, region of interest (ROI) selection, ROI segmentation, radiomics mode, and algorithms.ResultsIn total, 20 studies classified as type 2a or above according to the Transparent Reporting of a Multivariable Prediction Model for Individual Prognosis or Diagnosis (TRIPOD) statement were included for this systematic review and meta-analysis. The studies achieved an average RQS of 13 (ranging from 10 to 17), accounting for 36.1% of the total points. The average METRICS score was 69.3% (ranging from 54.8% to 80.9%). The quality category of the studies is mainly "good". The results of our meta-analysis showed that the pooled sensitivity (SEN), specificity (SPE), positive likelihood ratio, negative likelihood ratio and diagnostic odds ratio with 95% confidence interval (CI) were 0.82 (0.78, 0.88), 0.83 (0.76, 0.87), 4.7 (3.4, 6.6), 0.20 (0.15, 0.28) and 23 (13, 42), respectively. The area under the curve of the summary receiver operating characteristic curve was 0.90 (0.87, 0.92). The results of Spearman correlation analysis revealed no threshold effect among the studies (P = .423). Significant heterogeneity was observed among the studies (overall I2 = 83.7%; 95% CI: 76.0, 88.9; P < .01). Meta-regressions indicated that sample size and ROI selection contributed to the heterogeneity in SEN, while algorithms affected the heterogeneity in SPE (P < .05). Deeks' test confirmed there was no significant publication bias in all the included studies. The Fagan nomogram showed an absolute increase of 34% in the post-test probability following a positive test result.ConclusionThe results supported that 18F-FDG PET/CT radiomics has high diagnostic value for lymphoma. However, there is high heterogeneity among different studies. In the future, clinical practicality needs to be substantiated by more prospective studies with rigorous adherence to existing guidelines and multicentric validation.
Collapse
Affiliation(s)
- Chaoying Liu
- Department of Medical Equipment, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
| | - Jun Zhao
- Department of Nuclear Medicine, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Heng Zhang
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
- Department of Radiotherapy, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| | - Xinye Ni
- Jiangsu Province Engineering Research Center of Medical Physics, Changzhou, Jiangsu, China
- Medical Physics Research Center, Nanjing Medical University, Changzhou, Jiangsu, China
- Key Laboratory of Medical Physics in Changzhou, Changzhou, Jiangsu, China
- Department of Radiotherapy, the Third Affiliated Hospital of Nanjing Medical University, The Second People's Hospital of Changzhou, Changzhou, Jiangsu, China
| |
Collapse
|
6
|
Pozzato C, Outeiro-Pinho G, Galiè M, Ramadori G, Konstantinidou G. ERK5 suppression overcomes FAK inhibitor resistance in mutant KRAS-driven non-small cell lung cancer. EMBO Mol Med 2024; 16:2402-2426. [PMID: 39271958 PMCID: PMC11473843 DOI: 10.1038/s44321-024-00138-7] [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: 11/02/2023] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Mutated KRAS serves as the oncogenic driver in 30% of non-small cell lung cancers (NSCLCs) and is associated with metastatic and therapy-resistant tumors. Focal Adhesion Kinase (FAK) acts as a mediator in sustaining KRAS-driven lung tumors, and although FAK inhibitors are currently undergoing clinical development, clinical data indicated that their efficacy in producing long-term anti-tumor responses is limited. Here we revealed two FAK interactors, extracellular-signal-regulated kinase 5 (ERK5) and cyclin-dependent kinase 5 (CDK5), as key players underlying FAK-mediated maintenance of KRAS mutant NSCLC. Inhibition of ERK5 and CDK5 synergistically suppressed FAK function, decreased proliferation and induced apoptosis owing to exacerbated ROS-induced DNA damage. Accordingly, concomitant pharmacological inhibition of ERK5 and CDK5 in a mouse model of KrasG12D-driven lung adenocarcinoma suppressed tumor progression and promoted cancer cell death. Cancer cells resistant to FAK inhibitors showed enhanced ERK5-FAK signaling dampening DNA damage. Notably, ERK5 inhibition prevented the development of resistance to FAK inhibitors, significantly enhancing the efficacy of anti-tumor responses. Therefore, we propose ERK5 inhibition as a potential co-targeting strategy to counteract FAK inhibitor resistance in NSCLC.
Collapse
Affiliation(s)
- Chiara Pozzato
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | | - Mirco Galiè
- Department of Neuroscience, Biomedicine and Movement, University of Verona, 37134, Verona, Italy
| | - Giorgio Ramadori
- Department of Cell Physiology and Metabolism, University of Geneva, 1211, Geneva, Switzerland
- Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva, Switzerland
| | | |
Collapse
|
7
|
Bhimani R, Thompson JD, Suh N, Kadakia RJ, Bariteau JT, Kerkhoffs GMMJ, Coleman MM. Weightbearing Computed Tomography Can Accurately Detect Subtle Lisfranc Injury. Foot Ankle Int 2024; 45:1145-1155. [PMID: 39080976 DOI: 10.1177/10711007241266844] [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: 10/26/2024]
Abstract
BACKGROUND Early detection of Lisfranc injury is critical for improving clinical outcomes, but diagnosing subtle injury can be difficult. Weightbearing computed tomography (WBCT) allows evaluation of such injuries in 3 dimensions (3D) under physiologic load. This study aimed to assess the utility of 1-, 2-, and 3-dimensional measurements on WBCT to diagnose subtle injury in isolated ligamentous Lisfranc injuries. METHODS Ten cadaveric specimens underwent WBCT evaluation of the Lisfranc joint complex in the intact state and subsequently with sequential sectioning of the dorsal Lisfranc ligament and interosseous Lisfranc ligament (IOL) to create subtle Lisfranc injury, and finally after transectioning of plantar Lisfranc ligament (PLL) to create the injury conditions for complete ligamentous Lisfranc injury. Measurements under static vertical tibial load of 80 kg were performed on WBCT images including (1) Lisfranc joint (medial cuneiform-base of second metatarsal) volume, (2) Lisfranc joint area, (3) C1-C2 intercuneiform area, (4) C1-M2 distance, (5) C1-C2 distance, (6) M1-M2 intermetatarsal distance, (7) first tarsometatarsal (TMT1) alignment, (8) second tarsometatarsal (TMT2) alignment, (9) TMT1 dorsal step-off distance, and (10) TMT2 dorsal step-off distance. RESULTS In the subtle Lisfranc injury state, Lisfranc joint volume and area, C1-M2 distance, and M1-M2 distance measurements on WBCT significantly increased, when compared with the intact state (P values .001 to .014). Additionally, Lisfranc joint volume and area, C1-M2 distance, M1-M2 distance, TMT2 alignment, and TMT2 dorsal step-off measurements were increased in the complete Lisfranc injury state. Of all measurements, C1-M2 distance had the largest area under the curve (AUC) of 0.96 (sensitivity = 90%; specificity = 90%), followed by Lisfranc volume (AUC = 0.90; sensitivity = 80%; specificity = 80%) and Lisfranc area (AUC = 0.89; sensitivity = 80%; specificity = 100%). CONCLUSION In a cadaveric model we found that WBCT scan can increase the diagnostic accuracy for subtle Lisfranc injury. Among the measurements, C1-M2 distance exhibited the highest level of accuracy. The 2D joint area and 3D joint volume also proved to be accurate, with 3D volume measurements of the Lisfranc joint displaying the most significant absolute difference between the intact state and increasing severity of Lisfranc injury. These findings suggest that 2D joint area and 3D joint volume may have potential as supplementary measurements to more accurately diagnose subtle Lisfranc injuries. CLINICAL RELEVANCE WBCT may help surgeons detect subtle Lisfranc injuries.
Collapse
Affiliation(s)
- Rohan Bhimani
- Foot & Ankle Service, Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
- Department of Orthopedic Surgery, Academic Medical Center, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - J Daniel Thompson
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
| | - Nina Suh
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
| | - Rishin J Kadakia
- Foot & Ankle Service, Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
| | - Jason T Bariteau
- Foot & Ankle Service, Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
| | - Gino M M J Kerkhoffs
- Department of Orthopedic Surgery, Academic Medical Center, University of Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Michelle M Coleman
- Foot & Ankle Service, Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
- Department of Orthopaedic Surgery, Emory University Hospital, Atlanta, GA, USA
| |
Collapse
|
8
|
Bernatz S, Reddin IG, Fenton TR, Vogl TJ, Wild PJ, Köllermann J, Mandel P, Wenzel M, Hoeh B, Mahmoudi S, Koch V, Grünewald LD, Hammerstingl R, Döring C, Harter PN, Weber KJ. Epigenetic profiling of prostate cancer reveals potential prognostic signatures. J Cancer Res Clin Oncol 2024; 150:396. [PMID: 39180680 PMCID: PMC11344710 DOI: 10.1007/s00432-024-05921-0] [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: 07/03/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE While epigenetic profiling discovered biomarkers in several tumor entities, its application in prostate cancer is still limited. We explored DNA methylation-based deconvolution of benign and malignant prostate tissue for biomarker discovery and the potential of radiomics as a non-invasive surrogate. METHODS We retrospectively included 30 patients (63 [58-79] years) with prostate cancer (PCa) who had a multiparametric MRI of the prostate before radical prostatectomy between 2014 and 2019. The control group comprised four patients with benign prostate tissue adjacent to the PCa lesions and four patients with benign prostatic hyperplasia. Tissue punches of all lesions were obtained. DNA methylation analysis and reference-free in silico deconvolution were conducted to retrieve Latent Methylation Components (LCMs). LCM-based clustering was analyzed for cellular composition and correlated with clinical disease parameters. Additionally, PCa and adjacent benign lesions were analyzed using radiomics to predict the epigenetic signatures non-invasively. RESULTS LCMs identified two clusters with potential prognostic impact. Cluster one was associated with malignant prostate tissue (p < 0.001) and reduced immune-cell-related signatures (p = 0.004) of CD19 and CD4 cells. Cluster one comprised exclusively malignant prostate tissue enriched for significant prostate cancer and advanced tumor stages (p < 0.03 for both). No radiomics model could non-invasively predict the epigenetic clusters. CONCLUSION Epigenetic clusters were associated with prognostically and clinically relevant metrics in prostate cancer. Further, immune cell-related signatures differed significantly between prognostically favorable and unfavorable clusters. Further research is necessary to explore potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Simon Bernatz
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
| | - Ian G Reddin
- School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Tim R Fenton
- School of Cancer Sciences, University of Southampton, Southampton, UK
| | - Thomas J Vogl
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Peter J Wild
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), Frankfurt am Main, Germany
| | - Jens Köllermann
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
| | - Philipp Mandel
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Mike Wenzel
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Benedikt Hoeh
- Goethe University Frankfurt, University Hospital, Department of Urology, Frankfurt am Main, Germany
| | - Scherwin Mahmoudi
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Vitali Koch
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Leon D Grünewald
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Renate Hammerstingl
- Goethe University Frankfurt, University Hospital, Clinic for Radiology and Nuclear Medicine, Frankfurt am Main, Germany
| | - Claudia Döring
- Goethe University Frankfurt, University Hospital, Dr. Senckenberg Institute for Pathology, Frankfurt am Main, Germany
| | - Patrick N Harter
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany
- Center for Neuropathology and Prion Research, Faculty of Medicine, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, A Partnership Between DKFZ and University/University Hospital, LMU Munich, Munich, Germany
| | - Katharina J Weber
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany.
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT), Frankfurt am Main, Germany.
| |
Collapse
|
9
|
Taton O, Van Muylem A, Leduc D, Gevenois PA. CT-Based Evaluation of the Shape of the Diaphragm Using 3D Slicer. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1980-1990. [PMID: 38467956 PMCID: PMC11300794 DOI: 10.1007/s10278-024-01069-y] [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: 01/11/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
The diaphragm is the main inspiratory muscle and separates the thorax and the abdomen. In COPD, the evaluation of the diaphragm shape is clinically important, especially in the case of hyperinflation. However, delineating the diaphragm remains a challenge as it cannot be seen entirely on CT scans. Therefore, the lungs, ribs, sternum, and lumbar vertebrae are used as surrogate landmarks to delineate the diaphragm. We herein describe a CT-based method for evaluating the shape of the diaphragm using 3D Slicer-a free software that allows delineation of the diaphragm landmarks-in ten COPD patients. Using the segmentation performed with 3D Slicer, the diaphragm shape was reconstructed with open-source Free Pascal Compiler. From this graduated model, the length of the muscle fibers, the radius of curvature, and the area of the diaphragm-the main determinants of its function-can be measured. Inter- and intra-user variabilities were evaluated with Bland and Altman plots and linear mixed models. Except for the coronal length (p = 0.049), there were not statistically significant inter- or intra-user differences (p values ranging from 0.326 to 0.910) suggesting that this method is reproducible and repeatable. In conclusion, 3D Slicer can be applied to CT scans for determining the shape of the diaphragm in COPD patients.
Collapse
Affiliation(s)
- Olivier Taton
- Department of Pneumology, Hôpital Erasme, Université libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium.
| | - Alain Van Muylem
- Department of Pneumology, Hôpital Erasme, Université libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Dimitri Leduc
- Department of Pneumology, Hôpital Erasme, Université libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium
| | - Pierre Alain Gevenois
- Department of Pneumology, Hôpital Erasme, Université libre de Bruxelles (ULB), 808 Route de Lennik, 1070, Brussels, Belgium
| |
Collapse
|
10
|
Gotta J, Gruenewald LD, Geyer T, Eichler K, Martin SS, Mahmoudi S, Booz C, Biciusca T, Reschke P, Juergens LJ, Sommer CM, D'Angelo T, Almansour H, Onay M, Herrmann E, Vogl TJ, Koch V. Indicators for Hospitalization in Acute Pulmonary Embolism: Uncover the Association Between D-dimer Levels, Thrombus Volume and Radiomics. Acad Radiol 2024; 31:2610-2619. [PMID: 38242733 DOI: 10.1016/j.acra.2023.12.045] [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: 12/17/2023] [Revised: 12/23/2023] [Accepted: 12/30/2023] [Indexed: 01/21/2024]
Abstract
BACKGROUND The advent of advanced computed tomography (CT) technology and the field of radiomics has opened up new avenues in diagnostic assessments. Increasingly, there is substantial evidence advocating for the incorporation of quantitative imaging biomarkers in the clinical decision-making process. This study aimed to examine the correlation between D-dimer levels and thrombus size in acute pulmonary embolism (PE) combining dual-energy CT (DECT) and radiomics and to investigate the diagnostic utility of a machine learning classifier based on dual-energy computed tomography (DECT) radiomics for identifying patients with a complicated course, defined as at least hospitalization at IMC. METHODS The study was conducted including 136 participants who underwent pulmonary artery CT angiography from January 2015 to March 2022. Based on DECT imaging, 107 radiomic features were extracted for each patient using standardized image processing. After dividing the dataset into training and test sets, stepwise feature reduction based on reproducibility, variable importance and correlation analyses were performed to select the most relevant features; these were used to train and validate the gradient-boosted tree models.Receiver operating characteristics (ROC) analysis was utilized to evaluate the association between volumetric, laboratory data and adverse outcomes. RESULTS In the central PE group, we observed a significant correlation between thrombus volumetrics and D-dimer levels (p = 0.0037), as well as between thrombus volumetrics and hospitalization at the Intermediate Care Unit (IMC) (p = 0.0001). In contrast, no statistically significant differences were identified in thrombus sizes between patients who experienced complications and those who had a favorable course (p = 0.3162). The trained machine learning classifier achieved an accuracy of 61% and 55% in identifying patients with a complicated course, as indicated by an area under the ROC curve of 0.63 and 0.58. CONCLUSION In conclusion, our findings indicate a positive correlation between D-dimer levels and central PE's pulmonary embolic burden. Thrombus volumetrics may serve as an indicator for complications and outcomes in acute PE patients. Thus, thrombus volumetrics, as opposed to D-dimers, could be an additional marker for evaluating embolic disease severity. Moreover, DECT-derived radiomic feature models show promise in identifying patients with a complicated course, such as hospitalization at IMC.
Collapse
Affiliation(s)
- Jennifer Gotta
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.).
| | - Leon D Gruenewald
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Tobias Geyer
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Katrin Eichler
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Simon S Martin
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Scherwin Mahmoudi
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Christian Booz
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Teodora Biciusca
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Philipp Reschke
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Lisa-Joy Juergens
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Christof M Sommer
- Clinic of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany (C.M.S.)
| | - Tommaso D'Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy (T.D.)
| | - Haidara Almansour
- Department of Diagnostic and Interventional Radiology, Tuebingen University Hospital, Tuebingen, Germany (H.A.)
| | - Melis Onay
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany (M.O.)
| | - Eva Herrmann
- Institute for Biostatistics and Mathematic Modelling, Goethe University Frankfurt, 60590, Frankfurt, Germany (E.H.)
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| | - Vitali Koch
- Goethe University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany (J.G., L.D.G., T.G., K.E., S.S.M., S.M., C.B., T.B., P.R., J.J., T.J.V., V.K.)
| |
Collapse
|
11
|
Gotta J, Gruenewald LD, Martin SS, Booz C, Mahmoudi S, Eichler K, Gruber-Rouh T, Biciusca T, Reschke P, Juergens LJ, Onay M, Herrmann E, Scholtz JE, Sommer CM, Vogl TJ, Koch V. From pixels to prognosis: Imaging biomarkers for discrimination and outcome prediction of pulmonary embolism : Original Research Article. Emerg Radiol 2024; 31:303-311. [PMID: 38523224 PMCID: PMC11130040 DOI: 10.1007/s10140-024-02216-2] [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: 12/13/2023] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE Recent advancements in medical imaging have transformed diagnostic assessments, offering exciting possibilities for extracting biomarker-based information. This study aims to investigate the capabilities of a machine learning classifier that incorporates dual-energy computed tomography (DECT) radiomics. The primary focus is on discerning and predicting outcomes related to pulmonary embolism (PE). METHODS The study included 131 participants who underwent pulmonary artery DECT angiography between January 2015 and March 2022. Among them, 104 patients received the final diagnosis of PE and 27 patients served as a control group. A total of 107 radiomic features were extracted for every case based on DECT imaging. The dataset was divided into training and test sets for model development and validation. Stepwise feature reduction identified the most relevant features, which were used to train a gradient-boosted tree model. Receiver operating characteristics analysis and Cox regression tests assessed the association of texture features with overall survival. RESULTS The trained machine learning classifier achieved a classification accuracy of 0.94 for identifying patients with acute PE with an area under the receiver operating characteristic curve of 0.91. Radiomics features could be valuable for predicting outcomes in patients with PE, demonstrating strong prognostic capabilities in survival prediction (c-index, 0.991 [0.979-1.00], p = 0.0001) with a median follow-up of 130 days (IQR, 38-720). Notably, the inclusion of clinical or DECT parameters did not enhance predictive performance. CONCLUSION In conclusion, our study underscores the promising potential of leveraging radiomics on DECT imaging for the identification of patients with acute PE and predicting their outcomes. This approach has the potential to improve clinical decision-making and patient management, offering efficiencies in time and resources by utilizing existing DECT imaging without the need for an additional scoring system.
Collapse
Affiliation(s)
- Jennifer Gotta
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany.
- University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590, Germany.
| | | | - Simon S Martin
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christian Booz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Katrin Eichler
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Teodora Biciusca
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Philipp Reschke
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Melis Onay
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Eva Herrmann
- Institut for Biostatistics and Mathematic Modelling, Goethe University Frankfurt, Frankfurt, 60590, Germany
| | - Jan-Erik Scholtz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christof M Sommer
- Clinic of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Vitali Koch
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
12
|
Fu C, Dong J, Zhang J, Li X, Zuo S, Zhang H, Gao S, Chen L. Using three-dimensional model-based tumour volume change to predict the symptom improvement in patients with renal cell cancer. 3 Biotech 2024; 14:148. [PMID: 38711822 PMCID: PMC11070407 DOI: 10.1007/s13205-024-03967-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 02/26/2024] [Indexed: 05/08/2024] Open
Abstract
In our recent study, we explored the efficacy of three-dimensional (3D) measurement of tumor volume in predicting the improvement of quality of life (QoL) in patients suffering from renal cell cancer (RCC), who were treated with axitinib and anti-PD-L1 antibodies. This study encompassed 18 RCC patients, including 10 men and 8 women, with an average age of 56.83 ± 9.94 years. By utilizing 3D Slicer software, we analyzed pre- and post-treatment CT scans to assess changes in tumor volume. Patients' QoL was evaluated through the FKSI-DRS questionnaire. Our findings revealed that 3D models for all patients were successfully created, and there was a moderate agreement between treatment response classifications based on RECIST 1.1 criteria and volumetric analysis (kappa = 0.556, p = 0.001). Notably, nine patients reported a clinically meaningful improvement in QoL following the treatment. Interestingly, the change in tumor volume as indicated by the 3D model showed a higher area under the curve in predicting QoL improvement compared to the change in diameter measured by CT, although this difference was not statistically significant (z = 0.593, p = 0.553). Furthermore, a multivariable analysis identified the change in tumor volume based on the 3D model as an independent predictor of QoL improvement (odds ratio = 1.073, 95% CI 1.002-1.149, p = 0.045).In conclusion, our study suggests that the change in tumor volume measured by a 3D model may be a more effective predictor of symptom improvement in RCC patients than traditional CT-based diameter measurements. This offers a novel approach for assessing treatment response and patient well-being, presenting a significant advancement in the field of RCC treatment.
Collapse
Affiliation(s)
- ChengWei Fu
- Medical School of Chinese PLA, No. 28 Fuxing Road, Haidian District Beijing, 100853 China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039 China
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - JinKai Dong
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039 China
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - JingYun Zhang
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - XueChao Li
- Medical School of Chinese PLA, No. 28 Fuxing Road, Haidian District Beijing, 100853 China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039 China
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - ShiDong Zuo
- Medical School of Chinese PLA, No. 28 Fuxing Road, Haidian District Beijing, 100853 China
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039 China
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - HongTao Zhang
- Department of Radiology, The Fifth Medical Center, Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - Shen Gao
- Department of Radiology, The Fifth Medical Center, Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| | - LiJun Chen
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, No. 69 Yongding Road, Haidian District, Beijing, 100039 China
- Department of Urology, The Fifth Medical Center Chinese PLA General Hospital, No. Yard 8, Fengtai East Street, Beijing, 100071 China
| |
Collapse
|
13
|
Louis T, Lucia F, Cousin F, Mievis C, Jansen N, Duysinx B, Le Pennec R, Visvikis D, Nebbache M, Rehn M, Hamya M, Geier M, Salaun PY, Schick U, Hatt M, Coucke P, Lovinfosse P, Hustinx R. Identification of CT radiomic features robust to acquisition and segmentation variations for improved prediction of radiotherapy-treated lung cancer patient recurrence. Sci Rep 2024; 14:9028. [PMID: 38641673 PMCID: PMC11031577 DOI: 10.1038/s41598-024-58551-4] [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: 12/05/2023] [Accepted: 04/01/2024] [Indexed: 04/21/2024] Open
Abstract
The primary objective of the present study was to identify a subset of radiomic features extracted from primary tumor imaged by computed tomography of early-stage non-small cell lung cancer patients, which remain unaffected by variations in segmentation quality and in computed tomography image acquisition protocol. The robustness of these features to segmentation variations was assessed by analyzing the correlation of feature values extracted from lesion volumes delineated by two annotators. The robustness to variations in acquisition protocol was evaluated by examining the correlation of features extracted from high-dose and low-dose computed tomography scans, both of which were acquired for each patient as part of the stereotactic body radiotherapy planning process. Among 106 radiomic features considered, 21 were identified as robust. An analysis including univariate and multivariate assessments was subsequently conducted to estimate the predictive performance of these robust features on the outcome of early-stage non-small cell lung cancer patients treated with stereotactic body radiation therapy. The univariate predictive analysis revealed that robust features demonstrated superior predictive potential compared to non-robust features. The multivariate analysis indicated that linear regression models built with robust features displayed greater generalization capabilities by outperforming other models in predicting the outcomes of an external validation dataset.
Collapse
Affiliation(s)
- Thomas Louis
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
| | - François Lucia
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
- Radiation Oncology Department, University Hospital of Brest, Brest, France.
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France.
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Carole Mievis
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Nicolas Jansen
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Bernard Duysinx
- Division of Pulmonology, University Hospital of Liège, Liège, Belgium
| | - Romain Le Pennec
- Nuclear Medicine Department, University Hospital of Brest, Brest, France
- GETBO INSERM UMR 1304, University of Brest, UBO, Brest, France
| | | | - Malik Nebbache
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Martin Rehn
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Mohamed Hamya
- Radiation Oncology Department, University Hospital of Brest, Brest, France
| | - Margaux Geier
- Medical Oncology Department, University Hospital of Brest, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospital of Brest, Brest, France
- GETBO INSERM UMR 1304, University of Brest, UBO, Brest, France
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital of Brest, Brest, France
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, University of Brest, Brest, France
| | - Philippe Coucke
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
14
|
Gotta J, Koch V, Geyer T, Martin SS, Booz C, Mahmoudi S, Eichler K, Reschke P, D'Angelo T, Klimek K, Vogl TJ, Gruenewald LD. Imaging-based risk stratification of patients with pulmonary embolism based on dual-energy CT-derived radiomics. Eur J Clin Invest 2024; 54:e14139. [PMID: 38063028 DOI: 10.1111/eci.14139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/13/2023] [Accepted: 11/20/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND Technological progress in the acquisition of medical images and the extraction of underlying quantitative imaging data has introduced exciting prospects for the diagnostic assessment of a wide range of conditions. This study aims to investigate the diagnostic utility of a machine learning classifier based on dual-energy computed tomography (DECT) radiomics for classifying pulmonary embolism (PE) severity and assessing the risk for early death. METHODS Patients who underwent CT pulmonary angiogram (CTPA) between January 2015 and March 2022 were considered for inclusion in this study. Based on DECT imaging, 107 radiomic features were extracted for each patient using standardized image processing. After dividing the dataset into training and test sets, stepwise feature reduction based on reproducibility, variable importance and correlation analyses were performed to select the most relevant features; these were used to train and validate the gradient-boosted tree models. RESULTS The trained machine learning classifier achieved a classification accuracy of .90 for identifying high-risk PE patients with an area under the receiver operating characteristic curve of .59. This CT-based radiomics signature showed good diagnostic accuracy for risk stratification in individuals presenting with central PE, particularly within higher risk groups. CONCLUSION Models utilizing DECT-derived radiomics features can accurately stratify patients with pulmonary embolism into established clinical risk scores. This approach holds the potential to enhance patient management and optimize patient flow by assisting in the clinical decision-making process. It also offers the advantage of saving time and resources by leveraging existing imaging to eliminate the necessity for manual clinical scoring.
Collapse
Affiliation(s)
- Jennifer Gotta
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Vitali Koch
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tobias Geyer
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Simon S Martin
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Christian Booz
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | | - Katrin Eichler
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Philipp Reschke
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Tommaso D'Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Konrad Klimek
- Goethe University Frankfurt, University Hospital, Clinic for Nuclear Medicine, Frankfurt am Main, Germany
| | - Thomas J Vogl
- Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | | |
Collapse
|
15
|
Demetriou D, Lockhat Z, Brzozowski L, Saini KS, Dlamini Z, Hull R. The Convergence of Radiology and Genomics: Advancing Breast Cancer Diagnosis with Radiogenomics. Cancers (Basel) 2024; 16:1076. [PMID: 38473432 DOI: 10.3390/cancers16051076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/09/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Despite significant progress in the prevention, screening, diagnosis, prognosis, and therapy of breast cancer (BC), it remains a highly prevalent and life-threatening disease affecting millions worldwide. Molecular subtyping of BC is crucial for predictive and prognostic purposes due to the diverse clinical behaviors observed across various types. The molecular heterogeneity of BC poses uncertainties in its impact on diagnosis, prognosis, and treatment. Numerous studies have highlighted genetic and environmental differences between patients from different geographic regions, emphasizing the need for localized research. International studies have revealed that patients with African heritage are often diagnosed at a more advanced stage and exhibit poorer responses to treatment and lower survival rates. Despite these global findings, there is a dearth of in-depth studies focusing on communities in the African region. Early diagnosis and timely treatment are paramount to improving survival rates. In this context, radiogenomics emerges as a promising field within precision medicine. By associating genetic patterns with image attributes or features, radiogenomics has the potential to significantly improve early detection, prognosis, and diagnosis. It can provide valuable insights into potential treatment options and predict the likelihood of survival, progression, and relapse. Radiogenomics allows for visual features and genetic marker linkage that promises to eliminate the need for biopsy and sequencing. The application of radiogenomics not only contributes to advancing precision oncology and individualized patient treatment but also streamlines clinical workflows. This review aims to delve into the theoretical underpinnings of radiogenomics and explore its practical applications in the diagnosis, management, and treatment of BC and to put radiogenomics on a path towards fully integrated diagnostics.
Collapse
Affiliation(s)
- Demetra Demetriou
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Zarina Lockhat
- Department of Radiology, Faculty of Health Sciences, Steve Biko Academic Hospital, University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Luke Brzozowski
- Translational Research and Core Facilities, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Kamal S Saini
- Fortrea Inc., 8 Moore Drive, Durham, NC 27709, USA
- Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, Pretoria 0028, South Africa
| |
Collapse
|
16
|
Gotta J, Gruenewald LD, Martin SS, Booz C, Eichler K, Mahmoudi S, Rezazadeh CÖ, Reschke P, Biciusca T, Juergens L, Mader C, Hammerstingl R, Sommer CM, Vogl TJ, Koch V. Unmasking pancreatic cancer: Advanced biomedical imaging for its detection in native versus arterial dual‐energy computed tomography (DECT) scans. INTERNATIONAL JOURNAL OF IMAGING SYSTEMS AND TECHNOLOGY 2024; 34. [DOI: 10.1002/ima.23037] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/27/2024] [Indexed: 01/23/2025]
Abstract
AbstractThis study investigates the potential of a machine learning classifier using dual‐ energy computed tomography (DECT) radiomics to differentiate between malignant pancreatic lesions and normal pancreas tissue. A total of 100 patients who underwent third‐generation DECT between November 2018 and October 2022 were included, with 60 patients having pancreatic cancer and 40 normal pancreatic tissue. Radiomics features were extracted from non‐contrast and arterial‐enhanced DECT scans with stepwise feature reduction used to identify relevant features. Thetrained machine learning classifiers achieved a diagnostic accuracy of 0.97 in the arterial‐enhanced model and 0.88 in non‐contrast scans with sensitivities of 0.97 and 0.96, respectively. Areas under the curve were 0.97 (95% CI, 0.92–1.0, p < 0.001) and 0.96 (95% CI, 0.90–1.0, p < 0.001), respectively with no significant differences between both models (p= 0.52). This approach shows promise in enhancing pancreatic cancer detection and improving patient diagnoses, particulary in specific patient groups.
Collapse
Affiliation(s)
- Jennifer Gotta
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | | | - Simon S. Martin
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | - Christian Booz
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | - Katrin Eichler
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | | | | | - Philipp Reschke
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | | | | | - Christoph Mader
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | | | - Christof M. Sommer
- Clinic of Diagnostic and Interventional Radiology Heidelberg University Hospital Heidelberg Germany
| | - Thomas J. Vogl
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| | - Vitali Koch
- Goethe University Hospital Frankfurt Frankfurt am Main Germany
| |
Collapse
|
17
|
Lucia F, Louis T, Cousin F, Bourbonne V, Visvikis D, Mievis C, Jansen N, Duysinx B, Le Pennec R, Nebbache M, Rehn M, Hamya M, Geier M, Salaun PY, Schick U, Hatt M, Coucke P, Hustinx R, Lovinfosse P. Multicentric development and evaluation of [ 18F]FDG PET/CT and CT radiomic models to predict regional and/or distant recurrence in early-stage non-small cell lung cancer treated by stereotactic body radiation therapy. Eur J Nucl Med Mol Imaging 2024; 51:1097-1108. [PMID: 37987783 DOI: 10.1007/s00259-023-06510-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/03/2023] [Indexed: 11/22/2023]
Abstract
PURPOSE To develop machine learning models to predict regional and/or distant recurrence in patients with early-stage non-small cell lung cancer (ES-NSCLC) after stereotactic body radiation therapy (SBRT) using [18F]FDG PET/CT and CT radiomics combined with clinical and dosimetric parameters. METHODS We retrospectively collected 464 patients (60% for training and 40% for testing) from University Hospital of Liège and 63 patients from University Hospital of Brest (external testing set) with ES-NSCLC treated with SBRT between 2010 and 2020 and who had undergone pretreatment [18F]FDG PET/CT and planning CT. Radiomic features were extracted using the PyRadiomics toolbox®. The ComBat harmonization method was applied to reduce the batch effect between centers. Clinical, radiomic, and combined models were trained and tested using a neural network approach to predict regional and/or distant recurrence. RESULTS In the training (n = 273) and testing sets (n = 191 and n = 63), the clinical model achieved moderate performances to predict regional and/or distant recurrence with C-statistics from 0.53 to 0.59 (95% CI, 0.41, 0.67). The radiomic (original_firstorder_Entropy, original_gldm_LowGrayLevelEmphasis and original_glcm_DifferenceAverage) model achieved higher predictive ability in the training set and kept the same performance in the testing sets, with C-statistics from 0.70 to 0.78 (95% CI, 0.63, 0.88) while the combined model performs moderately well with C-statistics from 0.50 to 0.62 (95% CI, 0.37, 0.69). CONCLUSION Radiomic features extracted from pre-SBRT analog and digital [18F]FDG PET/CT outperform clinical parameters in the prediction of regional and/or distant recurrence and to discuss an adjuvant systemic treatment in ES-NSCLC. Prospective validation of our models should now be carried out.
Collapse
Affiliation(s)
- François Lucia
- Radiation Oncology Department, University Hospital, Brest, France.
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France.
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
- Service de Radiothérapie, CHRU Morvan, 2 Avenue Foch, 29609 Cedex, Brest, France.
| | - Thomas Louis
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - François Cousin
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | | | - Carole Mievis
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Nicolas Jansen
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | | | - Romain Le Pennec
- Nuclear Medicine Department, University Hospital, Brest, France
- GETBO, INSERM, UMR 1304, University of Brest, UBO, Brest, France
| | - Malik Nebbache
- Radiation Oncology Department, University Hospital, Brest, France
| | - Martin Rehn
- Radiation Oncology Department, University Hospital, Brest, France
| | - Mohamed Hamya
- Radiation Oncology Department, University Hospital, Brest, France
| | - Margaux Geier
- Medical Oncology Department, University Hospital, Brest, France
| | - Pierre-Yves Salaun
- Nuclear Medicine Department, University Hospital, Brest, France
- GETBO, INSERM, UMR 1304, University of Brest, UBO, Brest, France
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Philippe Coucke
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
- GIGA-CRC In Vivo Imaging, University of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
18
|
Wu ZX, Bu WQ, Tang Y, Guo YX, Guo YC, Wang F, Meng HT. Sex estimation using maxillary sinus volume for Chinese subjects based on cone-beam computed tomography. BMC Oral Health 2024; 24:253. [PMID: 38374033 PMCID: PMC10875788 DOI: 10.1186/s12903-024-04010-5] [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: 11/15/2023] [Accepted: 02/09/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Sex estimate is a key stage in forensic science for identifying individuals. Some anatomical structures may be useful for sex estimation since they retain their integrity even after highly severe events. However, few studies are focusing on the Chinese population. Some researchers used teeth for sex estimation, but comparison with maxillary sinus were lack. As a result, the objective of this research is to develop a sex estimation formula for the northwestern Chinese population by the volume of the maxillary sinus and compare with the accuracy of sex estimation based on teeth through cone-beam computed tomography (CBCT). METHODS CBCT images from 349 samples were used to establish and verify the formula. The volume of both the left and right maxillary sinuses was measured and examined for appropriate formula coefficients. To create the formula, we randomly picked 80% of the data as the training set and 20% of the samples as the testing set. Another set of samples, including 20 males and 20 females, were used to compare the accuracy of maxillary sinuses and teeth. RESULTS Overall, sex estimation accuracy by volume of the left maxillary sinus can reach 78.57%, while by the volume of the right maxillary sinus can reach 74.29%. The accuracy for females, which can reach 91.43% using the left maxillary sinus, was significantly higher than that for males, which was 65.71%. The result also shows that maxillary sinus volume was higher in males. The comparison with the available results using measurements of teeth for sex estimation performed by our group showed that the accuracy of sex estimation using canines volume was higher than the one using maxillary sinus volume, the accuracies based on mesiodistal diameter of canine and first molar were the same or lower than the volume of maxillary sinus. CONCLUSIONS The study demonstrates that measurement of maxillary sinus volume based on CBCT scans was an available and alternative method for sex estimation. And we established a method to accurately assess the sex of the northwest Chinese population. The comparison with the results of teeth measurements made the conclusion more reliable.
Collapse
Affiliation(s)
- Zi-Xuan Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Wen-Qing Bu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Yu Tang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Yu-Xin Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Yu-Cheng Guo
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China
- College of Medicine and Forensics, Xi'an Jiaotong University Health Science Center, 76 West Yanta Road, Xi'an, 710004, Shaanxi, People's Republic of China
| | - Fei Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China.
- Department of Orthodontics, Stomatological Hospital of Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China.
| | - Hao-Tian Meng
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, 98 XiWu Road, Xi'an, 710004, Shaanxi, People's Republic of China.
| |
Collapse
|
19
|
Maino C, Vernuccio F, Cannella R, Franco PN, Giannini V, Dezio M, Pisani AR, Blandino AA, Faletti R, De Bernardi E, Ippolito D, Gatti M, Inchingolo R. Radiomics and liver: Where we are and where we are headed? Eur J Radiol 2024; 171:111297. [PMID: 38237517 DOI: 10.1016/j.ejrad.2024.111297] [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: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 02/10/2024]
Abstract
Hepatic diffuse conditions and focal liver lesions represent two of the most common scenarios to face in everyday radiological clinical practice. Thanks to the advances in technology, radiology has gained a central role in the management of patients with liver disease, especially due to its high sensitivity and specificity. Since the introduction of computed tomography (CT) and magnetic resonance imaging (MRI), radiology has been considered the non-invasive reference modality to assess and characterize liver pathologies. In recent years, clinical practice has moved forward to a quantitative approach to better evaluate and manage each patient with a more fitted approach. In this setting, radiomics has gained an important role in helping radiologists and clinicians characterize hepatic pathological entities, in managing patients, and in determining prognosis. Radiomics can extract a large amount of data from radiological images, which can be associated with different liver scenarios. Thanks to its wide applications in ultrasonography (US), CT, and MRI, different studies were focused on specific aspects related to liver diseases. Even if broadly applied, radiomics has some advantages and different pitfalls. This review aims to summarize the most important and robust studies published in the field of liver radiomics, underlying their main limitations and issues, and what they can add to the current and future clinical practice and literature.
Collapse
Affiliation(s)
- Cesare Maino
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy.
| | - Federica Vernuccio
- Institute of Radiology, University Hospital of Padova, Padova 35128, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Paolo Niccolò Franco
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Valentina Giannini
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Michele Dezio
- Department of Radiology, Miulli Hospital, Acquaviva delle Fonti 70021, Bari, Italy
| | - Antonio Rosario Pisani
- Nuclear Medicine Unit, Interdisciplinary Department of Medicine, University of Bari, Bari 70121, Italy
| | - Antonino Andrea Blandino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Riccardo Faletti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Elisabetta De Bernardi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4, University of Milano Bicocca, Milano 20100, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Davide Ippolito
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Marco Gatti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Riccardo Inchingolo
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| |
Collapse
|
20
|
Deng H, Ge H, Dubey C, Losmanova T, Medová M, Konstantinidou G, Mutlu SM, Birrer FE, Brodie TM, Stroka D, Wang W, Peng RW, Dorn P, Marti TM. An optimized protocol for the generation and monitoring of conditional orthotopic lung cancer in the KP mouse model using an adeno-associated virus vector compatible with biosafety level 1. Cancer Immunol Immunother 2023; 72:4457-4470. [PMID: 37796299 PMCID: PMC10700219 DOI: 10.1007/s00262-023-03542-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND The inducible Kras/p53 lung adenocarcinoma mouse model, which faithfully recapitulates human disease, is routinely initiated by the intratracheal instillation of a virus-based Cre recombinase delivery system. Handling virus-based delivery systems requires elevated biosafety levels, e.g., biosafety level 2 (BSL-2). However, in experimental animal research facilities, following exposure to viral vectors in a BSL-2 environment, rodents may not be reclassified to BSL-1 according to standard practice, preventing access to small animal micro-computed tomography (micro-CT) scanners that are typically housed in general access areas such as BSL-1 rooms. Therefore, our goal was to adapt the protocol so that the Cre-induced KP mouse model could be handled under BSL-1 conditions during the entire procedure. RESULTS The Kras-Lox-STOP-Lox-G12D/p53 flox/flox (KP)-based lung adenocarcinoma mouse model was activated by intratracheal instillation of either an adenoviral-based or a gutless, adeno-associated viral-based Cre delivery system. Tumor growth was monitored over time by micro-CT. We have successfully substituted the virus-based Cre delivery system with a commercially available, gutless, adeno-associated, Cre-expressing vector that allows the KP mouse model to be handled and imaged in a BSL-1 facility. By optimizing the anesthesia protocol and switching to a microscope-guided vector instillation procedure, productivity was increased and procedure-related complications were significantly reduced. In addition, repeated micro-CT analysis of individual animals allowed us to monitor tumor growth longitudinally, dramatically reducing the number of animals required per experiment. Finally, we documented the evolution of tumor volume for different doses, which revealed that individual tumor nodules induced by low-titer AAV-Cre transductions can be monitored over time by micro-CT. CONCLUSION Modifications to the anesthesia and instillation protocols increased the productivity of the original KP protocol. In addition, the switch to a gutless, adeno-associated, Cre-expressing vector allowed longitudinal monitoring of tumor growth under BSL-1 conditions, significantly reducing the number of animals required for an experiment, in line with the 3R principles.
Collapse
Affiliation(s)
- Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Thoracic Surgery Department 2, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Huixiang Ge
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Christelle Dubey
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Michaela Medová
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Seyran Mathilde Mutlu
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Fabienne Esther Birrer
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Tess Melinda Brodie
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Deborah Stroka
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Wenxiang Wang
- Thoracic Surgery Department 2, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan, China
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Murtenstrasse 28, 3008, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
21
|
Bai J, Ito T, Fujikawa T, Honda K, Kawashima Y, Watanabe H, Kurata N, Tsutsumi T. Three-dimensionally visualized ossification and mineralization process of the otic capsule in a postnatal developmental mouse. Laryngoscope Investig Otolaryngol 2023; 8:1036-1043. [PMID: 37621296 PMCID: PMC10446274 DOI: 10.1002/lio2.1090] [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: 05/12/2023] [Accepted: 05/29/2023] [Indexed: 08/26/2023] Open
Abstract
Objective We aimed to elucidate the ossification process of the otic capsule in postnatal C57BL/6 mice and depict the three-dimensional (3D) process of otoconial mineralization in vivo. Methods The otic capsules of C57BL/6 mice were stained with alizarin red and imaged/compared using micro-computed tomography on postnatal day (P) between P0 and P8, P10, P15, and P30 and 3-4 months old (P3-4Mo). We reconstructed 3D images of the otic capsule and otoconia and measured the bone mineral density using x-ray absorptiometry on each age. Results The 3D reconstructed otic capsule images revealed two ossification centers of the otic capsule at P0. One was observed around the ampulla of the superior semicircular canal and utricle, and the other was observed around the ampulla of the posterior semicircular canal. The cross-sectional views demonstrated that modiolar ossification developed from the base to the apex from P4 to P8. The inter-scalar septum ossified bidirectionally from the modiolus and bony otic capsule from P8 to P15. The mineralized otoconia were first detected in the utricle at P3 and saccular otoconia at P6. The density of the utricle and saccular otoconia showed different growth trends. Conclusion To the best of our knowledge, this is the first study to demonstrate the 3D appearance of the otic capsule and otoconia in different developmental stages of mice. We also revealed that modiolar and inter-scalar septal calcification is the final event in the cochlea and that it can be susceptible to pathological conditions (cochlear congenital malformations and hereditary vestibular diseases). The unique features of the ossification process and duration may explain these pathological conditions observed in humans. Level of Evidence 3.
Collapse
Affiliation(s)
- Jing Bai
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | - Taku Ito
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | - Taro Fujikawa
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | - Keiji Honda
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | | | - Hiroki Watanabe
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | - Natsuko Kurata
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| | - Takeshi Tsutsumi
- Department of OtolaryngologyTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
22
|
Libling WA, Korn R, Weiss GJ. Review of the use of radiomics to assess the risk of recurrence in early-stage non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:1575-1589. [PMID: 37577298 PMCID: PMC10413018 DOI: 10.21037/tlcr-23-5] [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: 01/03/2023] [Accepted: 06/13/2023] [Indexed: 08/15/2023]
Abstract
Background and Objective Radiomics is an emerging field of advanced image analysis that has shown promise as a non-invasive, companion diagnostic in predicting clinical outcomes and response assessments in solid tumors. Radiomics aims to extract high-content information from medical images not visible to the naked eye, especially in early-stage non-small cell lung cancer (NSCLC) patients. Although these patients are being identified by early detection programs, it remains unclear which patients would benefit from adjuvant treatment versus active surveillance. Having a radiomic signature(s) that could predict early recurrence would be beneficial. In this review, an overview of the basic radiomic approaches used to evaluate solid tumors on radiologic scans, including NSCLC is provided followed by a review of relevant literature that supports the use of radiomics to help predict tumor recurrence in early-stage NSCLC patients. Methods A review of the radiomic literature from 1985 to present focusing on the prediction of disease recurrence in early-stage NSCLC was conducted. PubMed database was searched using key terms for radiomics and NSCLC. A total of 41 articles were identified and 13 studies were considered suitable for inclusion based upon study population, patient number (n>50), use of well described radiomic methodologies, suitable model building features, and well-defined testing/training and validation where feasible. Key Content and Findings Examples of using radiomics in early-stage NSCLC patients will be presented, where disease free survival is a primary consideration. A summary of the findings demonstrates the importance of both the intratumor and peritumoral radiomic signals as a marker of outcomes. Conclusions The value of radiomic information for predicting disease recurrence in early-stage NSCLC patients is accumulating. However, overcoming several challenges along with the lack of prospective trials, has inhibited it use as a clinical decision-making support tool in early-stage NSCLC.
Collapse
Affiliation(s)
- William Adam Libling
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Ronald Korn
- Virginia G Piper Cancer Center at HonorHealth, Scottsdale, AZ, USA
| | | |
Collapse
|
23
|
Lucia F, Bourbonne V, Pleyers C, Dupré PF, Miranda O, Visvikis D, Pradier O, Abgral R, Mervoyer A, Classe JM, Rousseau C, Vos W, Hermesse J, Gennigens C, De Cuypere M, Kridelka F, Schick U, Hatt M, Hustinx R, Lovinfosse P. Multicentric development and evaluation of 18F-FDG PET/CT and MRI radiomics models to predict para-aortic lymph node involvement in locally advanced cervical cancer. Eur J Nucl Med Mol Imaging 2023; 50:2514-2528. [PMID: 36892667 DOI: 10.1007/s00259-023-06180-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/27/2023] [Indexed: 03/10/2023]
Abstract
PURPOSE To develop machine learning models to predict para-aortic lymph node (PALN) involvement in patients with locally advanced cervical cancer (LACC) before chemoradiotherapy (CRT) using 18F-FDG PET/CT and MRI radiomics combined with clinical parameters. METHODS We retrospectively collected 178 patients (60% for training and 40% for testing) in 2 centers and 61 patients corresponding to 2 further external testing cohorts with LACC between 2010 to 2022 and who had undergone pretreatment analog or digital 18F-FDG PET/CT, pelvic MRI and surgical PALN staging. Only primary tumor volumes were delineated. Radiomics features were extracted using the Radiomics toolbox®. The ComBat harmonization method was applied to reduce the batch effect between centers. Different prediction models were trained using a neural network approach with either clinical, radiomics or combined models. They were then evaluated on the testing and external validation sets and compared. RESULTS In the training set (n = 102), the clinical model achieved a good prediction of the risk of PALN involvement with a C-statistic of 0.80 (95% CI 0.71, 0.87). However, it performed in the testing (n = 76) and external testing sets (n = 30 and n = 31) with C-statistics of only 0.57 to 0.67 (95% CI 0.36, 0.83). The ComBat-radiomic (GLDZM_HISDE_PET_FBN64 and Shape_maxDiameter2D3_PET_FBW0.25) and ComBat-combined (FIGO 2018 and same radiomics features) models achieved very high predictive ability in the training set and both models kept the same performance in the testing sets, with C-statistics from 0.88 to 0.96 (95% CI 0.76, 1.00) and 0.85 to 0.92 (95% CI 0.75, 0.99), respectively. CONCLUSIONS Radiomic features extracted from pre-CRT analog and digital 18F-FDG PET/CT outperform clinical parameters in the decision to perform a para-aortic node staging or an extended field irradiation to PALN. Prospective validation of our models should now be carried out.
Collapse
Affiliation(s)
- François Lucia
- Radiation Oncology Department, University Hospital, Brest, France.
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France.
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium.
| | - Vincent Bourbonne
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Clémence Pleyers
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | | | - Omar Miranda
- Radiation Oncology Department, University Hospital, Brest, France
| | | | - Olivier Pradier
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Ronan Abgral
- Nuclear Medicine Department, University Hospital, Brest, France
- EA GETBO 3878, IFR 148, University of Brest, UBO, Brest, France
| | - Augustin Mervoyer
- Department of Radiation Oncology, Institut de Cancérologie de l'Ouest Centre René Gauducheau, Saint Herblain, France
| | - Jean-Marc Classe
- Department of Surgical Oncology, Institut de Cancérologie de l'Ouest Centre René Gauducheau, Saint Herblain, France
| | - Caroline Rousseau
- Université de Nantes, CNRS, Inserm, CRCINA, F-44000, Nantes, France
- ICO René Gauducheau, F-44800, Saint-Herblain, France
| | - Wim Vos
- Radiomics SA, Liège, Belgium
| | - Johanne Hermesse
- Department of Radiotherapy Oncology, University Hospital of Liège, Liège, Belgium
| | - Christine Gennigens
- Department of Medical Oncology, University Hospital of Liège, Liège, Belgium
| | | | - Frédéric Kridelka
- Department of Gynecology, University Hospital of Liège, Liège, Belgium
| | - Ulrike Schick
- Radiation Oncology Department, University Hospital, Brest, France
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Mathieu Hatt
- LaTIM, INSERM, UMR 1101, Univ Brest, Brest, France
| | - Roland Hustinx
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| | - Pierre Lovinfosse
- Division of Nuclear Medicine and Oncological Imaging, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
24
|
Shakir H, Aijaz B, Khan TMR, Hussain M. A deep learning-based cancer survival time classifier for small datasets. Comput Biol Med 2023; 160:106896. [PMID: 37150085 DOI: 10.1016/j.compbiomed.2023.106896] [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: 12/22/2022] [Revised: 03/07/2023] [Accepted: 04/09/2023] [Indexed: 05/09/2023]
Abstract
Cancer survival time prediction using Deep Learning (DL) has been an emerging area of research. However, non-availability of large-sized annotated medical imaging databases affects the training performance of DL models leading to their arguable usage in many clinical applications. In this research work, a neural network model is customized for small sample space to avoid data over-fitting for DL training. A set of prognostic radiomic features is selected through an iterative process using average of multiple dropouts which results in back-propagated gradients with low variance, thus increasing the network learning capability, reliable feature selection and better training over a small database. The proposed classifier is further compared with erasing feature selection method proposed in the literature for improved network training and with other well-known classifiers on small sample size. Achieved results which were statistically validated show efficient and improved classification of cancer survival time into three intervals of 6 months, between 6 months up to 2 years, and above 2 years; and has the potential to aid health care professionals in lung tumor evaluation for timely treatment and patient care.
Collapse
Affiliation(s)
- Hina Shakir
- Department of Software Engineering, Bahria University, 13-National Stadium Road Karachi, 75620, Pakistan.
| | - Bushra Aijaz
- Department of Electrical Engineering, Bahria University, 13-National Stadium Road Karachi, 75620, Pakistan.
| | - Tariq Mairaj Rasool Khan
- Department of Electrical and Power Engineering, Pakistan Navy Engineering College, National University of Science and Technology, Karachi, Pakistan.
| | - Muhammad Hussain
- Department of Electrical Engineering, Bahria University, 13-National Stadium Road Karachi, 75620, Pakistan.
| |
Collapse
|
25
|
Koch V, Weitzer N, Dos Santos DP, Gruenewald LD, Mahmoudi S, Martin SS, Eichler K, Bernatz S, Gruber-Rouh T, Booz C, Hammerstingl RM, Biciusca T, Rosbach N, Gökduman A, D’Angelo T, Finkelmeier F, Yel I, Alizadeh LS, Sommer CM, Cengiz D, Vogl TJ, Albrecht MH. Multiparametric detection and outcome prediction of pancreatic cancer involving dual-energy CT, diffusion-weighted MRI, and radiomics. Cancer Imaging 2023; 23:38. [PMID: 37072856 PMCID: PMC10114410 DOI: 10.1186/s40644-023-00549-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/17/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND The advent of next-generation computed tomography (CT)- and magnetic resonance imaging (MRI) opened many new perspectives in the evaluation of tumor characteristics. An increasing body of evidence suggests the incorporation of quantitative imaging biomarkers into clinical decision-making to provide mineable tissue information. The present study sought to evaluate the diagnostic and predictive value of a multiparametric approach involving radiomics texture analysis, dual-energy CT-derived iodine concentration (DECT-IC), and diffusion-weighted MRI (DWI) in participants with histologically proven pancreatic cancer. METHODS In this study, a total of 143 participants (63 years ± 13, 48 females) who underwent third-generation dual-source DECT and DWI between November 2014 and October 2022 were included. Among these, 83 received a final diagnosis of pancreatic cancer, 20 had pancreatitis, and 40 had no evidence of pancreatic pathologies. Data comparisons were performed using chi-square statistic tests, one-way ANOVA, or two-tailed Student's t-test. For the assessment of the association of texture features with overall survival, receiver operating characteristics analysis and Cox regression tests were used. RESULTS Malignant pancreatic tissue differed significantly from normal or inflamed tissue regarding radiomics features (overall P < .001, respectively) and iodine uptake (overall P < .001, respectively). The performance for the distinction of malignant from normal or inflamed pancreatic tissue ranged between an AUC of ≥ 0.995 (95% CI, 0.955-1.0; P < .001) for radiomics features, ≥ 0.852 (95% CI, 0.767-0.914; P < .001) for DECT-IC, and ≥ 0.690 (95% CI, 0.587-0.780; P = .01) for DWI, respectively. During a follow-up of 14 ± 12 months (range, 10-44 months), the multiparametric approach showed a moderate prognostic power to predict all-cause mortality (c-index = 0.778 [95% CI, 0.697-0.864], P = .01). CONCLUSIONS Our reported multiparametric approach allowed for accurate discrimination of pancreatic cancer and revealed great potential to provide independent prognostic information on all-cause mortality.
Collapse
Affiliation(s)
- Vitali Koch
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Nils Weitzer
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Daniel Pinto Dos Santos
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Leon D. Gruenewald
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Scherwin Mahmoudi
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Simon S. Martin
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Katrin Eichler
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Simon Bernatz
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Tatjana Gruber-Rouh
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Christian Booz
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Renate M. Hammerstingl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Teodora Biciusca
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Nicolas Rosbach
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Aynur Gökduman
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Tommaso D’Angelo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University Hospital Messina, Messina, Italy
| | - Fabian Finkelmeier
- Department of Internal Medicine, University Hospital Frankfurt, Frankfurt Am Main, Germany
| | - Ibrahim Yel
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Leona S. Alizadeh
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Christof M. Sommer
- Clinic of Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Duygu Cengiz
- Department of Radiology, University of Koc School of Medicine, Istanbul, Turkey
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| | - Moritz H. Albrecht
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, Frankfurt am Main, 60590 Germany
| |
Collapse
|
26
|
Kawazoe Y, Shiinoki T, Fujimoto K, Yuasa Y, Hirano T, Matsunaga K, Tanaka H. Investigation of the combination of intratumoral and peritumoral radiomic signatures for predicting epidermal growth factor receptor mutation in lung adenocarcinoma. J Appl Clin Med Phys 2023:e13980. [DOI: 10.1002/acm2.13980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Affiliation(s)
- Yusuke Kawazoe
- Department of Radiation Oncology Graduate School of Medicine Yamaguchi University Ube Japan
| | - Takehiro Shiinoki
- Department of Radiation Oncology Graduate School of Medicine Yamaguchi University Ube Japan
| | - Koya Fujimoto
- Department of Radiation Oncology Graduate School of Medicine Yamaguchi University Ube Japan
| | - Yuki Yuasa
- Department of Radiation Oncology Graduate School of Medicine Yamaguchi University Ube Japan
| | - Tsunahiko Hirano
- Department of Respiratory Medicine and Infectious Disease Graduate School of Medicine Yamaguchi University Ube Japan
| | - Kazuto Matsunaga
- Department of Respiratory Medicine and Infectious Disease Graduate School of Medicine Yamaguchi University Ube Japan
| | - Hidekazu Tanaka
- Department of Radiation Oncology Graduate School of Medicine Yamaguchi University Ube Japan
| |
Collapse
|
27
|
Kawazoe Y, Shiinoki T, Fujimoto K, Yuasa Y, Hirano T, Matsunaga K, Tanaka H. Comparison of the radiomics-based predictive models using machine learning and nomogram for epidermal growth factor receptor mutation status and subtypes in lung adenocarcinoma. Phys Eng Sci Med 2023; 46:395-403. [PMID: 36787023 DOI: 10.1007/s13246-023-01232-9] [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: 12/16/2021] [Accepted: 02/01/2023] [Indexed: 02/15/2023]
Abstract
The purpose of this study is to develop the predictive models for epidermal growth factor receptor (EGFR) mutation status and subtypes [exon 21-point mutation (L858R) and exon 19 deletion mutation (19Del)] and evaluate their clinical usefulness. Total 172 patients with lung adenocarcinoma were retrospectively analyzed. The analysis of variance and the least absolute shrinkage were used for feature selection from plain computed tomography images. Then, radiomic score (rad-score) was calculated for the training and test cohorts. Two machine learning (ML) models with 5-fold were applied to construct the predictive models with rad-score, clinical features, and the combination of rad-score and clinical features. The nomogram was developed using rad-score and clinical features. The prediction performance was evaluated by the area under the receiver operating characteristic curve (AUC). Finally, decision curve analysis (DCA) was performed using the best ML and nomogram models. In the test cohorts, the AUC of the best ML and the nomogram model were 0.73 (95% confidence interval, 0.59-0.87) and 0.79 (0.65-0.92) in the EGFR mutation groups, 0.83 (0.67-0.99) and 0.85 (0.72-0.97) in the L858R mutation groups, as well as 0.77 (0.58-0.97) and 0.77 (0.60-0.95) in the 19Del groups. The DCA showed that the nomogram models have comparable results with ML models. We constructed two predictive models for EGFR mutation status and subtypes. The nomogram models had comparable results to the ML models. Because the superiority of the performance of ML and nomogram models varied depending on the prediction groups, appropriate model selection is necessary.
Collapse
Affiliation(s)
- Yusuke Kawazoe
- Department of Radiation Oncology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takehiro Shiinoki
- Department of Radiation Oncology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan.
| | - Koya Fujimoto
- Department of Radiation Oncology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Yuki Yuasa
- Department of Radiation Oncology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Tsunahiko Hirano
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kazuto Matsunaga
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hidekazu Tanaka
- Department of Radiation Oncology, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
28
|
Bernatz S, Elenberger O, Ackermann J, Lenga L, Martin SS, Scholtz JE, Koch V, Grünewald LD, Herrmann Y, Kinzler MN, Stehle A, Koch I, Zeuzem S, Bankov K, Doering C, Reis H, Flinner N, Schulze F, Wild PJ, Hammerstingl R, Eichler K, Gruber-Rouh T, Vogl TJ, Dos Santos DP, Mahmoudi S. CT-radiomics and clinical risk scores for response and overall survival prognostication in TACE HCC patients. Sci Rep 2023; 13:533. [PMID: 36631548 PMCID: PMC9834236 DOI: 10.1038/s41598-023-27714-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
We aimed to identify hepatocellular carcinoma (HCC) patients who will respond to repetitive transarterial chemoembolization (TACE) to improve the treatment algorithm. Retrospectively, 61 patients (mean age, 65.3 years ± 10.0 [SD]; 49 men) with 94 HCC mRECIST target-lesions who had three consecutive TACE between 01/2012 and 01/2020 were included. Robust and non-redundant radiomics features were extracted from the 24 h post-embolization CT. Five different clinical TACE-scores were assessed. Seven different feature selection methods and machine learning models were used. Radiomics, clinical and combined models were built to predict response to TACE on a lesion-wise and patient-wise level as well as its impact on overall-survival prognostication. 29 target-lesions of 19 patients were evaluated in the test set. Response rates were 37.9% (11/29) on the lesion-level and 42.1% (8/19) on the patient-level. Radiomics top lesion-wise response prognostications was AUC 0.55-0.67. Clinical scores revealed top AUCs of 0.65-0.69. The best working model combined the radiomic feature LargeDependenceHighGrayLevelEmphasis and the clinical score mHAP_II_score_group with AUC = 0.70, accuracy = 0.72. We transferred this model on a patient-level to achieve AUC = 0.62, CI = 0.41-0.83. The two radiomics-clinical features revealed overall-survival prognostication of C-index = 0.67. In conclusion, a random forest model using the radiomic feature LargeDependenceHighGrayLevelEmphasis and the clinical mHAP-II-score-group seems promising for TACE response prognostication.
Collapse
Affiliation(s)
- Simon Bernatz
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany.
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute (FCI), 60590, Frankfurt am Main, Germany.
| | - Oleg Elenberger
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Jörg Ackermann
- Department of Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt am Main, Robert-Mayer-Str. 11-15, 60325, Frankfurt am Main, Germany
| | - Lukas Lenga
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Simon S Martin
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Jan-Erik Scholtz
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Vitali Koch
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Leon D Grünewald
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Yannis Herrmann
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Maximilian N Kinzler
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Angelika Stehle
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Ina Koch
- Department of Molecular Bioinformatics, Institute of Computer Science, Goethe University Frankfurt am Main, Robert-Mayer-Str. 11-15, 60325, Frankfurt am Main, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Katrin Bankov
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Claudia Doering
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Henning Reis
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Nadine Flinner
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Falko Schulze
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
| | - Peter J Wild
- Dr. Senckenberg Institute for Pathology, University Hospital Frankfurt, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60590, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies (FIAS), 60438, Frankfurt am Main, Germany
| | - Renate Hammerstingl
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Katrin Eichler
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Tatjana Gruber-Rouh
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Thomas J Vogl
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| | - Daniel Pinto Dos Santos
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
- Department of Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
| | - Scherwin Mahmoudi
- University Hospital Frankfurt, Department of Diagnostic and Interventional Radiology, Goethe University Frankfurt am Main, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Tsai HY, Tsai TY, Wu CH, Chung WS, Wang JC, Hsu JS, Hou MF, Chou MC. Integration of Clinical and CT-Based Radiomic Features for Pretreatment Prediction of Pathologic Complete Response to Neoadjuvant Systemic Therapy in Breast Cancer. Cancers (Basel) 2022; 14:cancers14246261. [PMID: 36551746 PMCID: PMC9777141 DOI: 10.3390/cancers14246261] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The purpose of the present study was to examine the potential of a machine learning model with integrated clinical and CT-based radiomics features in predicting pathologic complete response (pCR) to neoadjuvant systemic therapy (NST) in breast cancer. Contrast-enhanced CT was performed in 329 patients with breast tumors (n = 331) before NST. Pyradiomics was used for feature extraction, and 107 features of seven classes were extracted. Feature selection was performed on the basis of the intraclass correlation coefficient (ICC), and six ICC thresholds (0.7−0.95) were examined to identify the feature set resulting in optimal model performance. Clinical factors, such as age, clinical stage, cancer cell type, and cell surface receptors, were used for prediction. We tried six machine learning algorithms, and clinical, radiomics, and clinical−radiomics models were trained for each algorithm. Radiomics and clinical−radiomics models with gray level co-occurrence matrix (GLCM) features only were also built for comparison. The linear support vector machine (SVM) regression model trained with radiomics features of ICC ≥0.85 in combination with clinical factors performed the best (AUC = 0.87). The performance of the clinical and radiomics linear SVM models showed statistically significant difference after correction for multiple comparisons (AUC = 0.69 vs. 0.78; p < 0.001). The AUC of the radiomics model trained with GLCM features was significantly lower than that of the radiomics model trained with all seven classes of radiomics features (AUC = 0.85 vs. 0.87; p = 0.011). Integration of clinical and CT-based radiomics features was helpful in the pretreatment prediction of pCR to NST in breast cancer.
Collapse
Affiliation(s)
- Huei-Yi Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Tsung-Yu Tsai
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chia-Hui Wu
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Wei-Shiuan Chung
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Imaging, Kaohsiung Municipal Siaogang Hospital, Kaohsiung 812, Taiwan
| | - Jo-Ching Wang
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Jui-Sheng Hsu
- Department of Medical Imaging, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Feng Hou
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ming-Chung Chou
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Imaging and Radiological Science, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-312-1101 (ext. 2357-23)
| |
Collapse
|
30
|
Jha N, Kim T, Ham S, Baek SH, Sung SJ, Kim YJ, Kim N. Fully automated condyle segmentation using 3D convolutional neural networks. Sci Rep 2022; 12:20590. [PMID: 36446860 PMCID: PMC9709043 DOI: 10.1038/s41598-022-24164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/10/2022] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to develop an auto-segmentation algorithm for mandibular condyle using the 3D U-Net and perform a stress test to determine the optimal dataset size for achieving clinically acceptable accuracy. 234 cone-beam computed tomography images of mandibular condyles were acquired from 117 subjects from two institutions, which were manually segmented to generate the ground truth. Semantic segmentation was performed using basic 3D U-Net and a cascaded 3D U-Net. A stress test was performed using different sets of condylar images as the training, validation, and test datasets. Relative accuracy was evaluated using dice similarity coefficients (DSCs) and Hausdorff distance (HD). In the five stages, the DSC ranged 0.886-0.922 and 0.912-0.932 for basic 3D U-Net and cascaded 3D U-Net, respectively; the HD ranged 2.557-3.099 and 2.452-2.600 for basic 3D U-Net and cascaded 3D U-Net, respectively. Stage V (largest data from two institutions) exhibited the highest DSC of 0.922 ± 0.021 and 0.932 ± 0.023 for basic 3D U-Net and cascaded 3D U-Net, respectively. Stage IV (200 samples from two institutions) had a lower performance than stage III (162 samples from one institution). Our results show that fully automated segmentation of mandibular condyles is possible using 3D U-Net algorithms, and the segmentation accuracy increases as training data increases.
Collapse
Affiliation(s)
- Nayansi Jha
- Graduate School of Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Taehun Kim
- Department of Biomedical Engineering, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sungwon Ham
- Research Strategy Team, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung-Hak Baek
- Department of Orthodontics, School of Dentistry, Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Sang-Jin Sung
- Department of Orthodontics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon-Ji Kim
- Department of Orthodontics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Namkug Kim
- Department of Convergence Medicine, Asan Medical Center, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
31
|
Automatic lung tumor segmentation from CT images using improved 3D densely connected UNet. Med Biol Eng Comput 2022; 60:3311-3323. [DOI: 10.1007/s11517-022-02667-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
|
32
|
Horvat N, Miranda J, El Homsi M, Peoples JJ, Long NM, Simpson AL, Do RKG. A primer on texture analysis in abdominal radiology. Abdom Radiol (NY) 2022; 47:2972-2985. [PMID: 34825946 DOI: 10.1007/s00261-021-03359-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/18/2023]
Abstract
The number of publications on texture analysis (TA), radiomics, and radiogenomics has been growing exponentially, with abdominal radiologists aiming to build new prognostic or predictive biomarkers for a wide range of clinical applications including the use of oncological imaging to advance the field of precision medicine. TA is specifically concerned with the study of the variation of pixel intensity values in radiological images. Radiologists aim to capture pixel variation in radiological images to deliver new insights into tumor biology that cannot be derived from visual inspection alone. TA remains an active area of investigation and requires further standardization prior to its clinical acceptance and applicability. This review is for radiologists interested in this rapidly evolving field, who are thinking of performing research or want to better interpret results in this arena. We will review the main concepts in TA, workflow processes, and existing challenges and steps to overcome them, as well as look at publications in body imaging with external validation.
Collapse
Affiliation(s)
- Natally Horvat
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo, SP, Brazil
| | - Maria El Homsi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Jacob J Peoples
- School of Computing, Queen's University, Kingston, ON, Canada
| | - Niamh M Long
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Amber L Simpson
- School of Computing, Queen's University, Kingston, ON, Canada.,Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada
| | - Richard K G Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| |
Collapse
|
33
|
Reproducibility and Repeatability of Coronary Computed Tomography Angiography (CCTA) Image Segmentation in Detecting Atherosclerosis: A Radiomics Study. Diagnostics (Basel) 2022; 12:diagnostics12082007. [PMID: 36010355 PMCID: PMC9406887 DOI: 10.3390/diagnostics12082007] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/23/2022] Open
Abstract
Atherosclerosis is known as the leading factor in heart disease with the highest mortality rate among the Malaysian population. Usually, the gold standard for diagnosing atherosclerosis is by using the coronary computed tomography angiography (CCTA) technique to look for plaque within the coronary artery. However, qualitative diagnosis for noncalcified atherosclerosis is vulnerable to false-positive diagnoses, as well as inconsistent reporting between observers. In this study, we assess the reproducibility and repeatability of segmenting atherosclerotic lesions manually and semiautomatically in CCTA images to identify the most appropriate CCTA image segmentation method for radiomics analysis to quantitatively extract the atherosclerotic lesion. Thirty (30) CCTA images were taken retrospectively from the radiology image database of Hospital Canselor Tuanku Muhriz (HCTM), Kuala Lumpur, Malaysia. We extract 11,700 radiomics features which include the first-order, second-order and shape features from 180 times of image segmentation. The interest vessels were segmentized manually and semiautomatically using LIFEx (Version 7.0.15, Institut Curie, Orsay, France) software by two independent radiology experts, focusing on three main coronary blood vessels. As a result, manual segmentation with a soft-tissuewindowing setting yielded higher repeatability as compared to semiautomatic segmentation with a significant intraclass correlation coefficient (intra-CC) 0.961 for thefirst-order and shape features; intra-CC of 0.924 for thesecond-order features with p < 0.001. Meanwhile, the semiautomatic segmentation has higher reproducibility as compared to manual segmentation with significant interclass correlation coefficient (inter-CC) of 0.920 (first-order features) and a good interclass correlation coefficient of 0.839 for the second-order features with p < 0.001. The first-order, shape order and second-order features for both manual and semiautomatic segmentation have an excellent percentage of reproducibility and repeatability (intra-CC > 0.9). In conclusion, semi-automated segmentation is recommended for inter-observer study while manual segmentation with soft tissue-windowing can be used for single observer study.
Collapse
|
34
|
Xie Q, Chen Y, Hu Y, Zeng F, Wang P, Xu L, Wu J, Li J, Zhu J, Xiang M, Zeng F. Development and validation of a machine learning-derived radiomics model for diagnosis of osteoporosis and osteopenia using quantitative computed tomography. BMC Med Imaging 2022; 22:140. [PMID: 35941568 PMCID: PMC9358842 DOI: 10.1186/s12880-022-00868-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/26/2022] [Indexed: 12/01/2022] Open
Abstract
Background To develop and validate a quantitative computed tomography (QCT) based radiomics model for discriminating osteoporosis and osteopenia.
Methods A total of 635 patients underwent QCT were retrospectively included from November 2016 to November 2019. The patients with osteopenia or osteoporosis (N = 590) were divided into a training cohort (N = 414) and a test cohort (N = 176). Radiomics features were extracted from the QCT images of the third lumbar vertebra. Minimum redundancy and maximum relevance and least absolute shrinkage and selection operator were used for data dimensional reduction, features selection and radiomics model building. Multivariable logistic regression was applied to construct the combined clinical-radiomic model that incorporated radiomics signatures and clinical characteristics. The performance of the combined clinical-radiomic model was evaluated by the area under the curve of receiver operator characteristic curve (ROC–AUC), accuracy, specificity, sensitivity, positive predictive value, and negative predictive value. Results The patients with osteopenia or osteoporosis were randomly divided into training and test cohort with a ratio of 7:3. Six more predictive radiomics signatures, age, alkaline phosphatase and homocysteine were selected to construct the combined clinical-radiomic model for diagnosis of osteoporosis and osteopenia. The AUC of the combined clinical-radiomic model was 0.96 (95% confidence interval (CI), 0.95 to 0.98) in the training cohort and 0.96 (95% CI 0.92 to 1.00) in the test cohort, which were superior to the clinical model alone (training-AUC = 0.81, test-AUC = 0.79). The calibration curve demonstrated that the radiomics nomogram had good agreement between prediction and observation and decision curve analysis confirmed clinically useful. Conclusions The combined clinical-radiomic model that incorporates the radiomics score and clinical risk factors, can serve as a reliable and powerful tool for discriminating osteoporosis and osteopenia. Supplementary Information The online version contains supplementary material available at 10.1186/s12880-022-00868-5.
Collapse
Affiliation(s)
- Qianrong Xie
- Department of Clinical Research Center, Dazhou Central Hospital, No.56 Nanyuemiao Street, Tongchuan District, Dazhou, 635000, Sichuan, China.,Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, 610000, China
| | - Yue Chen
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610000, Sichuan, China
| | - Yimei Hu
- Department of Orthopedics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Fanwei Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, No.56 Nanyuemiao Street, Tongchuan District, Dazhou, 635000, Sichuan, China
| | - Pingxi Wang
- Department of Bone Disease, Dazhou Central Hospital, Dazhou, 635000, China
| | - Lin Xu
- Department of Medical Imaging, Dazhou Central Hospital, Dazhou, 635000, China
| | - Jianhong Wu
- Department of Bone Disease, Dazhou Central Hospital, Dazhou, 635000, China
| | - Jie Li
- Department of Clinical Research Center, Dazhou Central Hospital, No.56 Nanyuemiao Street, Tongchuan District, Dazhou, 635000, Sichuan, China
| | - Jing Zhu
- Department of Rheumatology and Immunology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No.32 First Ring Road West, Jinniu District, Chengdu, 610000, Sichuan, China.
| | - Ming Xiang
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610000, Sichuan, China. .,Department of Orthopedics, Sichuan Provincial Orthopedic Hospital, Chengdu, 610000, China.
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, No.56 Nanyuemiao Street, Tongchuan District, Dazhou, 635000, Sichuan, China. .,Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
35
|
Lin JX, Lin JP, Weng Y, Lv CB, Chen JH, Zhan CY, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Zhou WX, Zhang XJ, Zheng CH, Cai LS, Ma YB, Huang CM. Radiographical Evaluation of Tumor Immunosuppressive Microenvironment and Treatment Outcomes in Gastric Cancer: A Retrospective, Multicohort Study. Ann Surg Oncol 2022; 29:5022-5033. [PMID: 35532827 DOI: 10.1245/s10434-022-11499-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
|
36
|
Deng H, Gao Y, Trappetti V, Hertig D, Karatkevich D, Losmanova T, Urzi C, Ge H, Geest GA, Bruggmann R, Djonov V, Nuoffer JM, Vermathen P, Zamboni N, Riether C, Ochsenbein A, Peng RW, Kocher GJ, Schmid RA, Dorn P, Marti TM. Targeting lactate dehydrogenase B-dependent mitochondrial metabolism affects tumor initiating cells and inhibits tumorigenesis of non-small cell lung cancer by inducing mtDNA damage. Cell Mol Life Sci 2022; 79:445. [PMID: 35877003 PMCID: PMC9314287 DOI: 10.1007/s00018-022-04453-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/21/2022] [Accepted: 06/27/2022] [Indexed: 02/08/2023]
Abstract
Once considered a waste product of anaerobic cellular metabolism, lactate has been identified as a critical regulator of tumorigenesis, maintenance, and progression. The putative primary function of lactate dehydrogenase B (LDHB) is to catalyze the conversion of lactate to pyruvate; however, its role in regulating metabolism during tumorigenesis is largely unknown. To determine whether LDHB plays a pivotal role in tumorigenesis, we performed 2D and 3D in vitro experiments, utilized a conventional xenograft tumor model, and developed a novel genetically engineered mouse model (GEMM) of non-small cell lung cancer (NSCLC), in which we combined an LDHB deletion allele with an inducible model of lung adenocarcinoma driven by the concomitant loss of p53 (also known as Trp53) and expression of oncogenic KRAS (G12D) (KP). Here, we show that epithelial-like, tumor-initiating NSCLC cells feature oxidative phosphorylation (OXPHOS) phenotype that is regulated by LDHB-mediated lactate metabolism. We show that silencing of LDHB induces persistent mitochondrial DNA damage, decreases mitochondrial respiratory complex activity and OXPHOS, resulting in reduced levels of mitochondria-dependent metabolites, e.g., TCA intermediates, amino acids, and nucleotides. Inhibition of LDHB dramatically reduced the survival of tumor-initiating cells and sphere formation in vitro, which can be partially restored by nucleotide supplementation. In addition, LDHB silencing reduced tumor initiation and growth of xenograft tumors. Furthermore, we report for the first time that homozygous deletion of LDHB significantly reduced lung tumorigenesis upon the concomitant loss of Tp53 and expression of oncogenic KRAS without considerably affecting the animal's health status, thereby identifying LDHB as a potential target for NSCLC therapy. In conclusion, our study shows for the first time that LDHB is essential for the maintenance of mitochondrial metabolism, especially nucleotide metabolism, demonstrating that LDHB is crucial for the survival and proliferation of NSCLC tumor-initiating cells and tumorigenesis.
Collapse
Affiliation(s)
- Haibin Deng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | | | - Damian Hertig
- Department of Neuroradiology, University of Bern, Bern, Switzerland
- Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
| | - Darya Karatkevich
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Christian Urzi
- Department of Neuroradiology, University of Bern, Bern, Switzerland
- Institute of Clinical Chemistry, University Hospital Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Huixiang Ge
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Gerrit Adriaan Geest
- Interfaculty Bioinformatics Unit, Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Remy Bruggmann
- Interfaculty Bioinformatics Unit, Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | | | - Jean-Marc Nuoffer
- Department of Neuroradiology, University of Bern, Bern, Switzerland
- Department of Pediatric Endocrinology, Diabetology and Metabolism, University Children's Hospital of Bern, Bern, Switzerland
| | - Peter Vermathen
- Translational Imaging Center (TIC), Swiss Institute for Translational and Entrepreneurial Medicine, Bern, Switzerland
| | - Nicola Zamboni
- Institute for Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Carsten Riether
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Adrian Ochsenbein
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Gregor Jan Kocher
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Ralph Alexander Schmid
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| | - Thomas Michael Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland.
- Department for BioMedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
37
|
Fast-Track-Protocol for Optimization of Presurgical Planning in Acute Surgical Treatment of Acetabular Quadrilateral Plate Fractures Using 3D Printing Technology and Pre-Contoured Reconstruction Plates. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12073492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background. Preoperative planning and 3D printing can be used to treat pelvic bone fractures using pre-contoured surgical plates, in particular complex, comminuted fractures involving the acetabulum and quadrilateral plate. The aim of the study was to develop a Fast-Track-Protocol (fast track methodology) for creating 3D anatomical models, that could be used to shape surgical plates, using open-source software and budget 3D printers. Such a ‘low-budget’ approach would allow a hospital-based multidisciplinary team to carry out pre-surgical planning and treat complex pelvic fractures using 3D technology. Methods. The study included 5 patients with comminuted pelvic fractures. For each patient, CT (computed tomography) data were converted into two 3D models of the pelvis-injured side and mirrored model of the contralateral, uninjured hemipelvis. These models were 3D printed and used as templates to shape surgical plates. Results. A Fast-Track-Protocol was established and used to successfully treat 5 patients with complex, comminuted fractures of the pelvis. Conclusion. Using the Fast-Track-Protocol it was possible to prepare 3D printed models and patient-specific pre-contoured plates within 2 days of hospital admittance. Such an approach resulted in better surgical technique and shorter operative times, while incurring relatively low costs.
Collapse
|
38
|
Fusco R, Granata V, Grazzini G, Pradella S, Borgheresi A, Bruno A, Palumbo P, Bruno F, Grassi R, Giovagnoni A, Grassi R, Miele V, Barile A. Radiomics in medical imaging: pitfalls and challenges in clinical management. Jpn J Radiol 2022; 40:919-929. [PMID: 35344132 DOI: 10.1007/s11604-022-01271-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 12/21/2022]
Abstract
BACKGROUND Radiomics and radiogenomics are two words that recur often in language of radiologists, nuclear doctors and medical physicists especially in oncology field. Radiomics is the technique of medical images analysis to extract quantitative data that are not detected by human eye. METHODS This article is a narrative review on Radiomics in Medical Imaging. In particular, the review exposes the process, the limitations related to radiomics, and future prospects are discussed. RESULTS Several studies showed that radiomics is very promising. However, there were some critical issues: poor standardization and generalization of radiomics results, data-quality control, repeatability, reproducibility, database balancing and issues related to model overfitting. CONCLUSIONS Radiomics procedure should made considered all pitfalls and challenges to obtain robust and reproducible results that could be generalized in other patients cohort.
Collapse
Affiliation(s)
| | - Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale-IRCCS di Napoli", Naples, Italy.
| | - Giulia Grazzini
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Silvia Pradella
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Alessandra Borgheresi
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Alessandra Bruno
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Pierpaolo Palumbo
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Diagnostic Imaging, Area of Cardiovascular and Interventional Imaging, Abruzzo Health Unit 1, 67100, L'Aquila, Italy
| | - Federico Bruno
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| | - Roberta Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Division of Radiology, "Università Degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Andrea Giovagnoni
- Department of Clinical Special and Dental Sciences, School of Radiology, University Politecnica delle Marche, Ancona, Italy
| | - Roberto Grassi
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Division of Radiology, "Università Degli Studi della Campania Luigi Vanvitelli", Naples, Italy
| | - Vittorio Miele
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy
| | - Antonio Barile
- Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, via della Signora 2, 20122, Milan, Italy.,Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, 67100, L'Aquila, Italy
| |
Collapse
|
39
|
Hosseini SA, Shiri I, Hajianfar G, Bahadorzade B, Ghafarian P, Zaidi H, Ay MR. Synergistic impact of motion and acquisition/reconstruction parameters on 18 F-FDG PET radiomic features in non-small cell lung cancer: phantom and clinical studies. Med Phys 2022; 49:3783-3796. [PMID: 35338722 PMCID: PMC9322423 DOI: 10.1002/mp.15615] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/12/2022] [Accepted: 03/14/2022] [Indexed: 11/25/2022] Open
Abstract
Objectives This study is aimed at examining the synergistic impact of motion and acquisition/reconstruction parameters on 18F‐FDG PET image radiomic features in non‐small cell lung cancer (NSCLC) patients, and investigating the robustness of features performance in differentiating NSCLC histopathology subtypes. Methods An in‐house developed thoracic phantom incorporating lesions with different sizes was used with different reconstruction settings, including various reconstruction algorithms, number of subsets and iterations, full‐width at half‐maximum of post‐reconstruction smoothing filter and acquisition parameters, including injected activity and test–retest with and without motion simulation. To simulate motion, a special motor was manufactured to simulate respiratory motion based on a normal patient in two directions. The lesions were delineated semi‐automatically to extract 174 radiomic features. All radiomic features were categorized according to the coefficient of variation (COV) to select robust features. A cohort consisting of 40 NSCLC patients with adenocarcinoma (n = 20) and squamous cell carcinoma (n = 20) was retrospectively analyzed. Statistical analysis was performed to discriminate robust features in differentiating histopathology subtypes of NSCLC lesions. Results Overall, 29% of radiomic features showed a COV ≤5% against motion. Forty‐five percent and 76% of the features showed a COV ≤ 5% against the test–retest with and without motion in large lesions, respectively. Thirty‐three percent and 45% of the features showed a COV ≤ 5% against different reconstruction parameters with and without motion, respectively. For NSCLC histopathological subtype differentiation, statistical analysis showed that 31 features were significant (p‐value < 0.05). Two out of the 31 significant features, namely, the joint entropy of GLCM (AUC = 0.71, COV = 0.019) and median absolute deviation of intensity histogram (AUC = 0.7, COV = 0.046), were robust against the motion (same reconstruction setting). Conclusions Motion, acquisition, and reconstruction parameters significantly impact radiomic features, just as their synergies. Radiomic features with high predictive performance (statistically significant) in differentiating histopathological subtype of NSCLC may be eliminated due to non‐reproducibility.
Collapse
Affiliation(s)
- Seyyed Ali Hosseini
- Department of Medical physics and biomedical engineering, Tehran University of medical sciences, Tehran, Iran.,Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Isaac Shiri
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, Switzerland
| | - Ghasem Hajianfar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | | | - Pardis Ghafarian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.,PET/CT and cyclotron center, Masih Daneshvari hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva 4, Switzerland.,Geneva University Neurocenter, Geneva University, CH-1205, Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, 9700 RB, Groningen, Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, DK-500, Odense, Denmark
| | - Mohammad Reza Ay
- Department of Medical physics and biomedical engineering, Tehran University of medical sciences, Tehran, Iran.,Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
40
|
D'Arnese E, Donato GWD, Sozzo ED, Sollini M, Sciuto D, Santambrogio MD. On the Automation of Radiomics-Based Identification and Characterization of NSCLC. IEEE J Biomed Health Inform 2022; 26:2670-2679. [PMID: 35255001 DOI: 10.1109/jbhi.2022.3156984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Proper detection and accurate characterization of Non-Small Cell Lung Cancer (NSCLC) are an open challenge in the imaging field. Biomedical imaging is fundamental in lung cancer assessment and offers the possibility of calculating predictive biomarkers impacting patients' management. Within this context, radiomics, which consists of extracting quantitative features from digital images, shows encouraging results for clinical applications, but the sub-optimal standardization of the procedure and the lack of definitive results are still a concern in the field. For these reasons, this work proposes the design and development of LuCIFEx, a fully-automated pipeline for non-invasive in-vivo characterization of NSCLC, aiming to speed up the analysis process and enable an early diagnosis of the tumor.LuCIFEx pipeline relies on routinely acquired [18F]FDG-PET/CT images for the automatic segmentation of the cancer lesion, allowing the computation of accurate radiomic features, then employed for cancer characterization through Machine Learning algorithms. The proposed multi-stage segmentation process can identify the lesion with a mean accuracy of 94.2±5.0%. Finally, the proposed data analysis pipeline demonstrates the potential of PET/CT features for the automatic recognition of lung metastases and NSCLC histological subtypes, while highlighting the main current limitations of the radiomic approach.
Collapse
|
41
|
Zhovannik I, Bontempi D, Romita A, Pfaehler E, Primakov S, Dekker A, Bussink J, Traverso A, Monshouwer R. Segmentation Uncertainty Estimation as a Sanity Check for Image Biomarker Studies. Cancers (Basel) 2022; 14:cancers14051288. [PMID: 35267597 PMCID: PMC8909427 DOI: 10.3390/cancers14051288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 11/16/2022] Open
Abstract
Problem. Image biomarker analysis, also known as radiomics, is a tool for tissue characterization and treatment prognosis that relies on routinely acquired clinical images and delineations. Due to the uncertainty in image acquisition, processing, and segmentation (delineation) protocols, radiomics often lack reproducibility. Radiomics harmonization techniques have been proposed as a solution to reduce these sources of uncertainty and/or their influence on the prognostic model performance. A relevant question is how to estimate the protocol-induced uncertainty of a specific image biomarker, what the effect is on the model performance, and how to optimize the model given the uncertainty. Methods. Two non-small cell lung cancer (NSCLC) cohorts, composed of 421 and 240 patients, respectively, were used for training and testing. Per patient, a Monte Carlo algorithm was used to generate three hundred synthetic contours with a surface dice tolerance measure of less than 1.18 mm with respect to the original GTV. These contours were subsequently used to derive 104 radiomic features, which were ranked on their relative sensitivity to contour perturbation, expressed in the parameter η. The top four (low η) and the bottom four (high η) features were selected for two models based on the Cox proportional hazards model. To investigate the influence of segmentation uncertainty on the prognostic model, we trained and tested the setup in 5000 augmented realizations (using a Monte Carlo sampling method); the log-rank test was used to assess the stratification performance and stability of segmentation uncertainty. Results. Although both low and high η setup showed significant testing set log-rank p-values (p = 0.01) in the original GTV delineations (without segmentation uncertainty introduced), in the model with high uncertainty, to effect ratio, only around 30% of the augmented realizations resulted in model performance with p < 0.05 in the test set. In contrast, the low η setup performed with a log-rank p < 0.05 in 90% of the augmented realizations. Moreover, the high η setup classification was uncertain in its predictions for 50% of the subjects in the testing set (for 80% agreement rate), whereas the low η setup was uncertain only in 10% of the cases. Discussion. Estimating image biomarker model performance based only on the original GTV segmentation, without considering segmentation, uncertainty may be deceiving. The model might result in a significant stratification performance, but can be unstable for delineation variations, which are inherent to manual segmentation. Simulating segmentation uncertainty using the method described allows for more stable image biomarker estimation, selection, and model development. The segmentation uncertainty estimation method described here is universal and can be extended to estimate other protocol uncertainties (such as image acquisition and pre-processing).
Collapse
Affiliation(s)
- Ivan Zhovannik
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.B.); (R.M.)
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
- Department of Radiation Oncology, The Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands
- Correspondence:
| | - Dennis Bontempi
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
| | - Alessio Romita
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
| | - Elisabeth Pfaehler
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
- University Clinic Augsburg, 86156 Augsburg, Germany
| | - Sergey Primakov
- The D-Lab, Department of Precision Medicine, GROW—School for Oncology, Maastricht University, 6229 ER Maastricht, The Netherlands;
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
| | - Johan Bussink
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.B.); (R.M.)
| | - Alberto Traverso
- Department of Radiation Oncology (Maastro), School for Oncology (GROW), Maastricht University Medical Center, 6229 ET Maastricht, The Netherlands; (D.B.); (A.R.); (E.P.); (A.D.); (A.T.)
| | - René Monshouwer
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (J.B.); (R.M.)
| |
Collapse
|
42
|
Chen X, Tong X, Qiu Q, Sun F, Yin Y, Gong G, Xing L, Sun X. Radiomics Nomogram for Predicting Locoregional Failure in Locally Advanced Non-small Cell Lung Cancer Treated with Definitive Chemoradiotherapy. Acad Radiol 2022; 29 Suppl 2:S53-S61. [PMID: 33308945 DOI: 10.1016/j.acra.2020.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 02/08/2023]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a computed tomography (CT)-based radiomics nomogram for predicting locoregional failure (LRF) in patients with locally advanced non-small cell lung cancer (NSCLC) treated with definitive chemoradiotherapy (CRT). MATERIALS AND METHODS A total of 141 patients with locally advanced NSCLC treated with definitive CRT from January 2014 to December 2017 were included and divided into testing cohort (n = 100) and validation (n = 41) cohort. Radiomics features were extracted from pretreatment contrast enhanced CT. The least absolute shrinkage and selection operator logistic regression was processed to select predictive features from the testing cohort and constructed a radiomics signature. Clinical characteristics and the radiomics signature were analyzed using univariable and multivariate Cox regression. The radiomics nomogram was established with the radiomics signature and independent clinical factors. Harrell's C-index, calibration curves and decision curves were used to assess the performance of the radiomics nomogram. RESULTS The radiomics signature, which consisted of eight selected features, was an independent factor of LRF. The clinical predictors of LRF were the histologic type and clinical stage. The radiomics nomogram combined with the radiomics signature and clinical prognostic factors showed good performance with C-indexes of 0.796 (95% confidence interval [CI]: 0.709-0.883) and 0.756 (95% CI: 0.674-0.838) in the testing and validation cohorts respectively. Additionally, the combined nomogram resulted in better performance (p < 0.001) for the estimation of LRF than the nomograms with the radiomics signature (C-index: 0.776; 95% CI: 0.686-0.866) or clinical predictors (C-index: 0.641; 95% CI: 0.542-0.740) alone. CONCLUSION The radiomics nomogram provided the best performance for LRF prediction in patients with locally advanced NSCLC, which may help optimize individual treatments.
Collapse
|
43
|
Kim DG, Park ES, Nam SM, Cha HG, Choi CY. Volumetric Evaluation of Dead Space in Ischial Pressure Injuries Using Magnetic Resonance Imaging: A Case Series. Adv Skin Wound Care 2021; 34:668-673. [PMID: 34807898 DOI: 10.1097/01.asw.0000797960.52759.75] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To establish a preoperative evaluation procedure by measuring the volume of dead space using MRI in patients with ischial pressure injuries. METHODS Patients with spinal cord injury and ischial pressure injuries who underwent treatment between August 2016 and November 2019 were included in the study. Preoperative MRI scan was conducted on all patients. The volume estimation and three-dimensional (3D) reconstruction were performed based on MRI data using a 3D Slicer. Based on the resulting volume, a muscle flap that could fit the dead space was selected. Surgery was performed with the selected muscle flap, and a fasciocutaneous flap was added, if necessary. RESULTS A total of eight patients with ischial pressure injuries were included in the study. The mean patient age was 59.0 ± 11.0 years. The mean body mass index was 26.62 ± 3.89 kg/m2. The mean volume of dead space was 104.75 ± 81.05 cm3. The gracilis muscle was the most selected muscle flap and was used in four patients. In five of eight cases, a fasciocutaneous flap was used as well. The mean follow-up period was 16 months, and by that point, none of the patients evinced complications that required surgery. CONCLUSIONS To the authors' knowledge, this is the first report on volumetric evaluation of dead space in ischial pressure injuries. The authors believe that the 3D reconstruction process would enable adequate dead space obliteration in ischial pressure injuries. The authors propose that preoperative MRI scans in patients with ischial pressure injury should become an essential part of the process.
Collapse
Affiliation(s)
- Dong Gyu Kim
- In the Department of Plastic and Reconstructive Surgery at the Soonchunhyang University Bucheon Hospital in Bucheon, Republic of Korea, Dong Gyu Kim, MD, is Resident; Eun Soo Park, MD, PhD, is Professor and Chief of the Medical Department; Seung Min Nam, MD, PhD, and Chang Yong Choi, MD, PhD are Associate Professors; and Han Gyu Cha, MD, is Assistant Professor. Acknowledgments : This work was supported by the Soonchunhyang University Research Fund. The authors have disclosed no other financial relationships related to this article. Submitted October 16, 2020; accepted in revised form January 26, 2021
| | | | | | | | | |
Collapse
|
44
|
Zhang G, Yang Z, Huo B, Chai S, Jiang S. Automatic segmentation of organs at risk and tumors in CT images of lung cancer from partially labelled datasets with a semi-supervised conditional nnU-Net. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 211:106419. [PMID: 34563895 DOI: 10.1016/j.cmpb.2021.106419] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/12/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Accurately and reliably defining organs at risk (OARs) and tumors are the cornerstone of radiation therapy (RT) treatment planning for lung cancer. Almost all segmentation networks based on deep learning techniques rely on fully annotated data with strong supervision. However, existing public imaging datasets encountered in the RT domain frequently include singly labelled tumors or partially labelled organs because annotating full OARs and tumors in CT images is both rigorous and tedious. To utilize labelled data from different sources, we proposed a dual-path semi-supervised conditional nnU-Net for OARs and tumor segmentation that is trained on a union of partially labelled datasets. METHODS The framework employs the nnU-Net as the base model and introduces a conditioning strategy by incorporating auxiliary information as an additional input layer into the decoder. The conditional nnU-Net efficiently leverages prior conditional information to classify the target class at the pixelwise level. Specifically, we employ the uncertainty-aware mean teacher (UA-MT) framework to assist in OARs segmentation, which can effectively leverage unlabelled data (images from a tumor labelled dataset) by encouraging consistent predictions of the same input under different perturbations. Furthermore, we individually design different combinations of loss functions to optimize the segmentation of OARs (Dice loss and cross-entropy loss) and tumors (Dice loss and focal loss) in a dual path. RESULTS The proposed method is evaluated on two publicly available datasets of the spinal cord, left and right lung, heart, esophagus, and lung tumor, in which satisfactory segmentation performance has been achieved in term of both the region-based Dice similarity coefficient (DSC) and the boundary-based Hausdorff distance (HD). CONCLUSIONS The proposed semi-supervised conditional nnU-Net breaks down the barriers between nonoverlapping labelled datasets and further alleviates the problem of "data hunger" and "data waste" in multi-class segmentation. The method has the potential to help radiologists with RT treatment planning in clinical practice.
Collapse
Affiliation(s)
- Guobin Zhang
- School of Mechanical Engineering, Tianjin University, Tianjin, 300350, China
| | - Zhiyong Yang
- School of Mechanical Engineering, Tianjin University, Tianjin, 300350, China
| | - Bin Huo
- Department of Oncology, Tianjin Medical University Second Hospital, Tianjin, 300211, China
| | - Shude Chai
- Department of Oncology, Tianjin Medical University Second Hospital, Tianjin, 300211, China
| | - Shan Jiang
- School of Mechanical Engineering, Tianjin University, Tianjin, 300350, China.
| |
Collapse
|
45
|
Ito T, Fujikawa T, Honda K, Makabe A, Watanabe H, Bai J, Kawashima Y, Miwa T, Griffith AJ, Tsutsumi T. Cochlear Pathomorphogenesis of Incomplete Partition Type II in Slc26a4-Null Mice. J Assoc Res Otolaryngol 2021; 22:681-691. [PMID: 34622375 DOI: 10.1007/s10162-021-00812-4] [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: 11/03/2020] [Accepted: 08/09/2021] [Indexed: 11/24/2022] Open
Abstract
Incomplete partition type II (IP-II) is frequently identified in ears with SLC26A4 mutations. Cochleae with IP-II are generally observed to have 1½ turns; the basal turns are normally formed, and the apical turn is dilated or cystic. The objective of this study was to characterize the pathomorphogenesis of the IP-II cochlear anomaly in Slc26a4-null mice. Otic capsules were dissected from Slc26a4Δ/+ and Slc26a4Δ/Δ mice at 1 and 8 days of age and at 1 and 3 months of age. X-ray micro-computed tomography was used to image samples. We used a multiplanar view and three-dimensional reconstructed models to calculate the cochlear duct length, cochlear turn rotation angle, and modiolus tilt angle. The number of inner hair cells was counted, and the length of the cochlear duct was measured in a whole-mount preparation of the membranous labyrinth. X-ray micro-computed tomography mid-modiolar planar views demonstrated cystic apical turns in Slc26a4Δ/Δ mice resulting from the loss or deossification of the interscalar septum, which morphologically resembles IP-II in humans. Planes vertical to the modiolus showed a similar mean rotation angle between Slc26a4Δ/+ and Slc26a4Δ/Δ mice. In contrast, the mean cochlear duct length and mean number of inner hair cells in Slc26a4Δ/Δ mice were significantly smaller than in Slc26a4Δ/+ mice. In addition, there were significant differences in the mean tilt angle and mean width of the modiolus. Our analysis of Slc26a4-null mice suggests that IP-II in humans reflects loss or deossification of the interscalar septum but not a decreased number of cochlear turns.
Collapse
Affiliation(s)
- Taku Ito
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan.
| | - Taro Fujikawa
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Keiji Honda
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Ayane Makabe
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Hiroki Watanabe
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Jing Bai
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Yoshiyuki Kawashima
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| | - Toru Miwa
- Department of Otolaryngology, Tazuke Kofukai Medical Research Institute, Kitano Hospital, 2-4-20 Ogimachi, Kita-ku, Osaka, 530-8480, Japan
| | - Andrew J Griffith
- Molecular Biology and Genetics Section, National Institute On Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, 20892, USA.,Departments of Otolaryngology and Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Takeshi Tsutsumi
- Department of Otorhinolaryngology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, 113-8519, Tokyo, Japan
| |
Collapse
|
46
|
Saadi SB, Ranjbarzadeh R, Ozeir kazemi, Amirabadi A, Ghoushchi SJ, Kazemi O, Azadikhah S, Bendechache M. Osteolysis: A Literature Review of Basic Science and Potential Computer-Based Image Processing Detection Methods. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2021; 2021:4196241. [PMID: 34646317 PMCID: PMC8505126 DOI: 10.1155/2021/4196241] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 09/14/2021] [Indexed: 12/22/2022]
Abstract
Osteolysis is one of the most prominent reasons of revision surgeries in total joint arthroplasty. This biological phenomenon is induced by wear particles and corrosion products that stimulate inflammatory biological response of surrounding tissues. The eventual responses of osteolysis are the activation of macrophages leading to bone resorption and prosthesis failure. Various factors are involved in the initiation of osteolysis from biological issues, design, material specifications, and model of the prosthesis to the health condition of the patient. Nevertheless, the factors leading to osteolysis are sometimes preventable. Changes in implant design and polyethylene manufacturing are striving to improve overall wear. Osteolysis is clinically asymptomatic and can be diagnosed and analyzed during follow-up sessions through various imaging modalities and methods, such as serial radiographic, CT scan, MRI, and image processing-based methods, especially with the use of artificial neural network algorithms. Deep learning algorithms with a variety of neural network structures such as CNN, U-Net, and Seg-UNet have proved to be efficient algorithms for medical image processing specifically in the field of orthopedics for the detection and segmentation of tumors. These deep learning algorithms can effectively detect and analyze osteolytic lesions well in advance during follow-up sessions in order to administer proper treatments before reaching a critical point. Osteolysis can be treated surgically or nonsurgically with medications. However, revision surgeries are the only solution for the progressive osteolysis. In this literature review, the underlying causes, mechanisms, and treatments of osteolysis are discussed with the main focus on the possible computer-based methods and algorithms that can be effectively employed for the detection of osteolysis.
Collapse
Affiliation(s)
- Soroush Baseri Saadi
- Department of Electrical Engineering, Islamic Azad University, South Tehran Branch, Tehran, Iran
| | - Ramin Ranjbarzadeh
- Department of Telecommunications Engineering, Faculty of Engineering, University of Guilan, Rasht, Iran
| | - Ozeir kazemi
- PPD - Global Pharmaceutical Contract Research Organization, Central Lab, Zaventem, Belgium
| | - Amir Amirabadi
- Department of Electrical Engineering, Islamic Azad University, South Tehran Branch, Tehran, Iran
| | | | | | - Sonya Azadikhah
- R.E.D. Laboratories N.V./S.A., Z.1 Researchpark, Zellik, Belgium
| | - Malika Bendechache
- School of Computing, Faculty of Engineering and Computing, Dublin City University, Dublin, Ireland
| |
Collapse
|
47
|
Sager P, Näf L, Vu E, Fischer T, Putora PM, Ehret F, Fürweger C, Schröder C, Förster R, Zwahlen DR, Muacevic A, Windisch P. Convolutional Neural Networks for Classifying Laterality of Vestibular Schwannomas on Single MRI Slices-A Feasibility Study. Diagnostics (Basel) 2021; 11:1676. [PMID: 34574017 PMCID: PMC8465488 DOI: 10.3390/diagnostics11091676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/04/2021] [Accepted: 09/07/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction: Many proposed algorithms for tumor detection rely on 2.5/3D convolutional neural networks (CNNs) and the input of segmentations for training. The purpose of this study is therefore to assess the performance of tumor detection on single MRI slices containing vestibular schwannomas (VS) as a computationally inexpensive alternative that does not require the creation of segmentations. Methods: A total of 2992 T1-weighted contrast-enhanced axial slices containing VS from the MRIs of 633 patients were labeled according to tumor location, of which 2538 slices from 539 patients were used for training a CNN (ResNet-34) to classify them according to the side of the tumor as a surrogate for detection and 454 slices from 94 patients were used for internal validation. The model was then externally validated on contrast-enhanced and non-contrast-enhanced slices from a different institution. Categorical accuracy was noted, and the results of the predictions for the validation set are provided with confusion matrices. Results: The model achieved an accuracy of 0.928 (95% CI: 0.869-0.987) on contrast-enhanced slices and 0.795 (95% CI: 0.702-0.888) on non-contrast-enhanced slices from the external validation cohorts. The implementation of Gradient-weighted Class Activation Mapping (Grad-CAM) revealed that the focus of the model was not limited to the contrast-enhancing tumor but to a larger area of the cerebellum and the cerebellopontine angle. Conclusions: Single-slice predictions might constitute a computationally inexpensive alternative to training 2.5/3D-CNNs for certain detection tasks in medical imaging even without the use of segmentations. Head-to-head comparisons between 2D and more sophisticated architectures could help to determine the difference in accuracy, especially for more difficult tasks.
Collapse
Affiliation(s)
- Philipp Sager
- Department of Radiation Oncology, Kantonsspital Winterthur, 8400 Winterthur, Switzerland; (P.S.); (C.S.); (R.F.); (D.R.Z.)
| | - Lukas Näf
- Department of Radiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland; (L.N.); (T.F.)
| | - Erwin Vu
- Department of Radiation Oncology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland; (E.V.); (P.M.P.)
| | - Tim Fischer
- Department of Radiology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland; (L.N.); (T.F.)
| | - Paul M. Putora
- Department of Radiation Oncology, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland; (E.V.); (P.M.P.)
- Department of Radiation Oncology, University of Bern, 3010 Bern, Switzerland
| | - Felix Ehret
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, 13353 Berlin, Germany;
- European Cyberknife Center, 81377 Munich, Germany; (C.F.); (A.M.)
| | - Christoph Fürweger
- European Cyberknife Center, 81377 Munich, Germany; (C.F.); (A.M.)
- Department of Stereotaxy and Functional Neurosurgery, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany
| | - Christina Schröder
- Department of Radiation Oncology, Kantonsspital Winterthur, 8400 Winterthur, Switzerland; (P.S.); (C.S.); (R.F.); (D.R.Z.)
| | - Robert Förster
- Department of Radiation Oncology, Kantonsspital Winterthur, 8400 Winterthur, Switzerland; (P.S.); (C.S.); (R.F.); (D.R.Z.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Daniel R. Zwahlen
- Department of Radiation Oncology, Kantonsspital Winterthur, 8400 Winterthur, Switzerland; (P.S.); (C.S.); (R.F.); (D.R.Z.)
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | | | - Paul Windisch
- Department of Radiation Oncology, Kantonsspital Winterthur, 8400 Winterthur, Switzerland; (P.S.); (C.S.); (R.F.); (D.R.Z.)
- European Cyberknife Center, 81377 Munich, Germany; (C.F.); (A.M.)
| |
Collapse
|
48
|
Sharma A, Tarbox L, Kurc T, Bona J, Smith K, Kathiravelu P, Bremer E, Saltz JH, Prior F. PRISM: A Platform for Imaging in Precision Medicine. JCO Clin Cancer Inform 2021; 4:491-499. [PMID: 32479186 PMCID: PMC7328100 DOI: 10.1200/cci.20.00001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Precision medicine requires an understanding of individual variability, which can only be acquired from large data collections such as those supported by the Cancer Imaging Archive (TCIA). We have undertaken a program to extend the types of data TCIA can support. This, in turn, will enable TCIA to play a key role in precision medicine research by collecting and disseminating high-quality, state-of-the-art, quantitative imaging data that meet the evolving needs of the cancer research community. METHODS A modular technology platform is presented that would allow existing data resources, such as TCIA, to evolve into a comprehensive data resource that meets the needs of users engaged in translational research for imaging-based precision medicine. This Platform for Imaging in Precision Medicine (PRISM) helps streamline the deployment and improve TCIA's efficiency and sustainability. More importantly, its inherent modular architecture facilitates a piecemeal adoption by other data repositories. RESULTS PRISM includes services for managing radiology and pathology images and features and associated clinical data. A semantic layer is being built to help users explore diverse collections and pool data sets to create specialized cohorts. PRISM includes tools for image curation and de-identification. It includes image visualization and feature exploration tools. The entire platform is distributed as a series of containerized microservices with representational state transfer interfaces. CONCLUSION PRISM is helping modernize, scale, and sustain the technology stack that powers TCIA. Repositories can take advantage of individual PRISM services such as de-identification and quality control. PRISM is helping scale image informatics for cancer research at a time when the size, complexity, and demands to integrate image data with other precision medicine data-intensive commons are mounting.
Collapse
Affiliation(s)
| | - Lawrence Tarbox
- University of Arkansas for Medical Sciences, Little Rock, AR
| | | | - Jonathan Bona
- University of Arkansas for Medical Sciences, Little Rock, AR
| | - Kirk Smith
- University of Arkansas for Medical Sciences, Little Rock, AR
| | | | | | | | - Fred Prior
- University of Arkansas for Medical Sciences, Little Rock, AR
| |
Collapse
|
49
|
Sakai K. [2. Radiomics of MRI]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2021; 77:866-875. [PMID: 34421076 DOI: 10.6009/jjrt.2021_jsrt_77.8.866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Koji Sakai
- Department of Radiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine
| |
Collapse
|
50
|
Kinsey CM, Billatos E, Mori V, Tonelli B, Cole BF, Duan F, Marques H, de la Bruere I, Onieva J, San José Estépar R, Cleveland A, Idelkope D, Stevenson C, Bates JHT, Aberle D, Spira A, Washko G, San José Estépar R. A simple assessment of lung nodule location for reduction in unnecessary invasive procedures. J Thorac Dis 2021; 13:4207-4216. [PMID: 34422349 PMCID: PMC8339782 DOI: 10.21037/jtd-20-3093] [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: 11/20/2020] [Accepted: 04/23/2021] [Indexed: 12/05/2022]
Abstract
Background CT screening for lung cancer results in a significant mortality reduction but is complicated by invasive procedures performed for evaluation of the many detected benign nodules. The purpose of this study was to evaluate measures of nodule location within the lung as predictors of malignancy. Methods We analyzed images and data from 3,483 participants in the National Lung Screening Trial (NLST). All nodules (4–20 mm) were characterized by 3D geospatial location using a Cartesian coordinate system and evaluated in logistic regression analysis. Model development and probability cutpoint selection was performed in the NLST testing set. The Geospatial test was then validated in the NLST testing set, and subsequently replicated in a new cohort of 147 participants from The Detection of Early Lung Cancer Among Military Personnel (DECAMP) Consortium. Results The Geospatial Test, consisting of the superior-inferior distance (Z distance), nodule diameter, and radial distance (carina to nodule) performed well in both the NLST validation set (AUC 0.85) and the DECAMP replication cohort (AUC 0.75). A negative Geospatial Test resulted in a less than 2% risk of cancer across all nodule diameters. The Geospatial Test correctly reclassified 19.7% of indeterminate nodules with a diameter over 6mm as benign, while only incorrectly classifying 1% of cancerous nodules as benign. In contrast, the parsimonious Brock Model applied to the same group of nodules correctly reclassified 64.5% of indeterminate nodules as benign but resulted in misclassification of a cancer as benign in 18.2% of the cases. Applying the Geospatial test would result in reducing invasive procedures performed for benign lesions by 11.3% with a low rate of misclassification (1.3%). In contrast, the Brock model applied to the same group of patients results in decreasing invasive procedures for benign lesion by 39.0% but misclassifying 21.1% of cancers as benign. Conclusions Utilizing information about geospatial location within the lung improves risk assessment for indeterminate lung nodules and may reduce unnecessary procedures. Trial Registration NCT00047385, NCT01785342.
Collapse
Affiliation(s)
- C Matthew Kinsey
- Division of Pulmonary and Critical Care, University of Vermont Medical Center, Burlington, VT, USA
| | - Ehab Billatos
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA, Boston Medical Center, Boston, MA, USA
| | - Vitor Mori
- University of Sao Paolo, Sao Paolo, Brazil
| | | | - Bernard F Cole
- Department of Mathematics and Statistics, University of Vermont, Burlington, VT, USA
| | - Fenghai Duan
- Department of Biostatistics and Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, USA
| | - Helga Marques
- Center for Statistical Sciences, Brown University School of Public Health, Providence, RI, USA
| | | | - Jorge Onieva
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | - Dan Idelkope
- Geisel School of Medicine at Dartmouth College, Hanover, NH, USA
| | | | - Jason H T Bates
- Division of Pulmonary and Critical Care, University of Vermont Medical Center, Burlington, VT, USA
| | - Denise Aberle
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Avi Spira
- The Pulmonary Unit, Boston Medical Center, Boston, MA, USA
| | - George Washko
- Division of Pulmonary and Critical Care, Brigham and Women's Hospital, Boston, MA, USA
| | | |
Collapse
|