Pub Date : 2024-01-01DOI: 10.1177/15330338231223384
{"title":"Erratum to \"Integrating Therapeutic Ultrasound With Nanosized Drug Delivery Systems in the Battle Against Cancer\".","authors":"","doi":"10.1177/15330338231223384","DOIUrl":"10.1177/15330338231223384","url":null,"abstract":"","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338231223384"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10860455/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139724032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241233443
Yifan Tang, Shuai Chen, Saijun Wang, Ke Xu, Kun Zhang, Dongmei Wang, Ninghan Feng
Purpose: Treatment of triple-negative breast cancer (TNBC) remains challenging. Intermittent fasting (IF) has emerged as a promising approach to improve metabolic health of various metabolic disorders. Clinical studies indicate IF is essential for TNBC progression. However, the molecular mechanisms underlying metabolic remodeling in regulating IF and TNBC progression are still unclear. Methods: In this study, we utilized a robust mouse model of TNBC and exposed subjects to a high-fat diet (HFD) with IF to explore its impact on the metabolic reprogramming linked to cancer progression. To identify crucial serum metabolites and signaling events, we utilized targeted metabolomics and RNA sequencing (RNA-seq). Furthermore, we conducted immunoblotting, real-time quantitative polymerase chain reaction (RT-qPCR), cell migration assays, lentivirus-mediated Mmp9 overexpression, and Mmp9 inhibitor experiments to elucidate the role of decanoylcarnitine/Mmp9 in TNBC cell migration. Results: Our observations indicate that IF exerts notable inhibitory effects on both the proliferation and cancer metastasis. Utilizing targeted metabolomics and RNA-seq, we initially identified pivotal serum metabolites and signaling events in the progression of TNBC. Among the 349 serum metabolites identified, decanoylcarnitine was picked out to inhibit TNBC cell proliferation and migration. RNA-seq analysis of TNBC cells treated with decanoylcarnitine revealed its suppressive effects on extracellular matrix-related protein components, with a notable reduction observed in Mmp9. Further investigations confirmed that decanoylcarnitine could inhibit Mmp9 expression in TNBC cells, primary tumors, lung, and liver metastasis tissues. Mmp9 overexpression abolished the inhibitory effect of decanoylcarnitine on cell migration. Conclusion: This study pioneers the exploration of IF intervention and the role of decanoylcarnitine/Mmp9 in the progression of TNBC in obese mice, enhancing our comprehension of the potential roles of various dietary patterns in the process of cancer treatment.
{"title":"Decanoylcarnitine Inhibits Triple-Negative Breast Cancer Progression via Mmp9 in an Intermittent Fasting Obesity Mouse.","authors":"Yifan Tang, Shuai Chen, Saijun Wang, Ke Xu, Kun Zhang, Dongmei Wang, Ninghan Feng","doi":"10.1177/15330338241233443","DOIUrl":"10.1177/15330338241233443","url":null,"abstract":"<p><p><b>Purpose:</b> Treatment of triple-negative breast cancer (TNBC) remains challenging. Intermittent fasting (IF) has emerged as a promising approach to improve metabolic health of various metabolic disorders. Clinical studies indicate IF is essential for TNBC progression. However, the molecular mechanisms underlying metabolic remodeling in regulating IF and TNBC progression are still unclear. <b>Methods:</b> In this study, we utilized a robust mouse model of TNBC and exposed subjects to a high-fat diet (HFD) with IF to explore its impact on the metabolic reprogramming linked to cancer progression. To identify crucial serum metabolites and signaling events, we utilized targeted metabolomics and RNA sequencing (RNA-seq). Furthermore, we conducted immunoblotting, real-time quantitative polymerase chain reaction (RT-qPCR), cell migration assays, lentivirus-mediated Mmp9 overexpression, and Mmp9 inhibitor experiments to elucidate the role of decanoylcarnitine/Mmp9 in TNBC cell migration. <b>Results:</b> Our observations indicate that IF exerts notable inhibitory effects on both the proliferation and cancer metastasis. Utilizing targeted metabolomics and RNA-seq, we initially identified pivotal serum metabolites and signaling events in the progression of TNBC. Among the 349 serum metabolites identified, decanoylcarnitine was picked out to inhibit TNBC cell proliferation and migration. RNA-seq analysis of TNBC cells treated with decanoylcarnitine revealed its suppressive effects on extracellular matrix-related protein components, with a notable reduction observed in Mmp9. Further investigations confirmed that decanoylcarnitine could inhibit Mmp9 expression in TNBC cells, primary tumors, lung, and liver metastasis tissues. Mmp9 overexpression abolished the inhibitory effect of decanoylcarnitine on cell migration. <b>Conclusion:</b> This study pioneers the exploration of IF intervention and the role of decanoylcarnitine/Mmp9 in the progression of TNBC in obese mice, enhancing our comprehension of the potential roles of various dietary patterns in the process of cancer treatment.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241233443"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10898300/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139973564","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241234790
Kareem A Attallah, Mohamed S Albannan, Khaled Farid, Sherine M Rizk, Nevine Fathy
Background: Hepatocellular carcinoma is frequently diagnosed in advanced stages, leading to a poorer prognosis. Therefore, early diagnosis and identification of biomarkers may significantly improve outcomes. Methods: This cross-sectional study enrolled 486 participants distributed among 3 groups: F1 to F3 = 184, F4 = 183, and hepatocellular carcinoma = 119. Liver fibrosis staging was performed using FibroScan, while imaging features were used for hepatocellular carcinoma detection. Epithelial membrane antigen and cytokeratin-1 levels in serum were quantified through Western blot and ELISA, respectively. Results: Patients diagnosed with hepatocellular carcinoma exhibited significantly elevated levels of epithelial membrane antigen and cytokeratin-1 compared to non-hepatocellular carcinoma patients, with a highly significant statistical difference (P < .0001). Epithelial membrane antigen demonstrated diagnostic performance with an area under the curve of 0.75, a sensitivity of 69.0%, and a specificity of 68.5%. Cytokeratin-1 for the identification of hepatocellular carcinoma showed a sensitivity of 79.0% and a specificity of 81.4%, resulting in an area under the curve of 0.87. The developed HCC-Check, which incorporates epithelial membrane antigen, cytokeratin-1, albumin, and alpha-fetoprotein, displayed a higher area under the curve of 0.95 to identify hepatocellular carcinoma, with a sensitivity of 89.8% and a specificity of 83.9%. Notably, HCC-Check values exceeding 2.57 substantially increased the likelihood of hepatocellular carcinoma, with an estimated odds ratio of 50.65, indicating a higher susceptibility to hepatocellular carcinoma development than those with lower values. The HCC-Check diagnostic test exhibited high precision in identifying patients with hepatocellular carcinoma, particularly those with small tumor sizes (<5 cm) and a single nodule, as reflected in area under the curve values of 0.92 and 0.85, respectively. HCC-Check was then applied to the validation study to test its accuracy and reproducibility, showing superior area under the curves for identifying different stages of hepatocellular carcinoma. These outcomes underscore the effectiveness of the test in the early detection of hepatocellular carcinoma. Conclusion: The HCC-Check test presents a highly accurate diagnostic method for detecting hepatocellular carcinoma in its early stages.
{"title":"HCC-Check: A Novel Diagnostic Tool for Early Detection of Hepatocellular Carcinoma Based on Cytokeratin-1 and Epithelial Membrane Antigen: A Cross-Sectional Study.","authors":"Kareem A Attallah, Mohamed S Albannan, Khaled Farid, Sherine M Rizk, Nevine Fathy","doi":"10.1177/15330338241234790","DOIUrl":"10.1177/15330338241234790","url":null,"abstract":"<p><p><b>Background:</b> Hepatocellular carcinoma is frequently diagnosed in advanced stages, leading to a poorer prognosis. Therefore, early diagnosis and identification of biomarkers may significantly improve outcomes. <b>Methods:</b> This cross-sectional study enrolled 486 participants distributed among 3 groups: F1 to F3 = 184, F4 = 183, and hepatocellular carcinoma = 119. Liver fibrosis staging was performed using FibroScan, while imaging features were used for hepatocellular carcinoma detection. Epithelial membrane antigen and cytokeratin-1 levels in serum were quantified through Western blot and ELISA, respectively. <b>Results:</b> Patients diagnosed with hepatocellular carcinoma exhibited significantly elevated levels of epithelial membrane antigen and cytokeratin-1 compared to non-hepatocellular carcinoma patients, with a highly significant statistical difference (<i>P </i>< .0001). Epithelial membrane antigen demonstrated diagnostic performance with an area under the curve of 0.75, a sensitivity of 69.0%, and a specificity of 68.5%. Cytokeratin-1 for the identification of hepatocellular carcinoma showed a sensitivity of 79.0% and a specificity of 81.4%, resulting in an area under the curve of 0.87. The developed HCC-Check, which incorporates epithelial membrane antigen, cytokeratin-1, albumin, and alpha-fetoprotein, displayed a higher area under the curve of 0.95 to identify hepatocellular carcinoma, with a sensitivity of 89.8% and a specificity of 83.9%. Notably, HCC-Check values exceeding 2.57 substantially increased the likelihood of hepatocellular carcinoma, with an estimated odds ratio of 50.65, indicating a higher susceptibility to hepatocellular carcinoma development than those with lower values. The HCC-Check diagnostic test exhibited high precision in identifying patients with hepatocellular carcinoma, particularly those with small tumor sizes (<5 cm) and a single nodule, as reflected in area under the curve values of 0.92 and 0.85, respectively. HCC-Check was then applied to the validation study to test its accuracy and reproducibility, showing superior area under the curves for identifying different stages of hepatocellular carcinoma. These outcomes underscore the effectiveness of the test in the early detection of hepatocellular carcinoma. <b>Conclusion:</b> The HCC-Check test presents a highly accurate diagnostic method for detecting hepatocellular carcinoma in its early stages.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241234790"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10913511/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140022589","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Early detection and accurate differentiation of malignant ground-glass nodules (GGNs) in lung CT scans are crucial for the effective treatment of lung adenocarcinoma. However, existing imaging diagnostic methods often struggle to distinguish between benign and malignant GGNs in the early stages. This study aims to predict the malignancy risk of GGNs observed in lung CT scans by applying two radiomics methods: topological data analysis and texture analysis.
Methods: A retrospective analysis was conducted on 3223 patients from two centers between January 2018 and June2023. The dataset was divided into training, testing, and validation sets to ensure robust model development and validation. We developed topological features applied to GGNs using radiomics analysis based on homology. This innovative approach emphasizes the integration of topological information, capturing complex geometric and spatial relationships within GGNs. By combining machine learning and deep learning algorithms, we established a predictive model that integrates clinical parameters, previous radiomics features, and topological radiomics features.
Results: Incorporating topological radiomics into our model significantly enhanced the ability to distinguish between benign and malignant GGNs. The topological radiomics model achieved areas under the curve (AUC) of 0.85 and 0.862 in two independent validation sets, outperforming previous radiomics models. Furthermore, this model demonstrated higher sensitivity compared to models based solely on clinical parameters, with sensitivities of 80.7% in validation set 1 and 82.3% in validation set 2. The most comprehensive model, which combined clinical parameters, previous radiomics features, and topological radiomics features, achieved the highest AUC value of 0.879 across all datasets.
Conclusion: This study validates the potential of topological radiomics in improving the predictive performance for distinguishing between benign and malignant GGNs. By integrating topological features with previous radiomics and clinical parameters, our comprehensive model provides a more accurate and reliable basis for developing treatment strategies for patients with GGNs.
{"title":"The Value of Topological Radiomics Analysis in Predicting Malignant Risk of Pulmonary Ground-Glass Nodules: A Multi-Center Study.","authors":"Miaoyu Wang, Yuanhui Wei, Minghui Zhu, Hang Yu, Chaomin Guo, Zhigong Chen, Wenjia Shi, Jiabo Ren, Wei Zhao, Zhen Yang, Liang-An Chen","doi":"10.1177/15330338241287089","DOIUrl":"10.1177/15330338241287089","url":null,"abstract":"<p><strong>Background: </strong>Early detection and accurate differentiation of malignant ground-glass nodules (GGNs) in lung CT scans are crucial for the effective treatment of lung adenocarcinoma. However, existing imaging diagnostic methods often struggle to distinguish between benign and malignant GGNs in the early stages. This study aims to predict the malignancy risk of GGNs observed in lung CT scans by applying two radiomics methods: topological data analysis and texture analysis.</p><p><strong>Methods: </strong>A retrospective analysis was conducted on 3223 patients from two centers between January 2018 and June2023. The dataset was divided into training, testing, and validation sets to ensure robust model development and validation. We developed topological features applied to GGNs using radiomics analysis based on homology. This innovative approach emphasizes the integration of topological information, capturing complex geometric and spatial relationships within GGNs. By combining machine learning and deep learning algorithms, we established a predictive model that integrates clinical parameters, previous radiomics features, and topological radiomics features.</p><p><strong>Results: </strong>Incorporating topological radiomics into our model significantly enhanced the ability to distinguish between benign and malignant GGNs. The topological radiomics model achieved areas under the curve (AUC) of 0.85 and 0.862 in two independent validation sets, outperforming previous radiomics models. Furthermore, this model demonstrated higher sensitivity compared to models based solely on clinical parameters, with sensitivities of 80.7% in validation set 1 and 82.3% in validation set 2. The most comprehensive model, which combined clinical parameters, previous radiomics features, and topological radiomics features, achieved the highest AUC value of 0.879 across all datasets.</p><p><strong>Conclusion: </strong>This study validates the potential of topological radiomics in improving the predictive performance for distinguishing between benign and malignant GGNs. By integrating topological features with previous radiomics and clinical parameters, our comprehensive model provides a more accurate and reliable basis for developing treatment strategies for patients with GGNs.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241287089"},"PeriodicalIF":2.7,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11452904/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142372935","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241227291
Yukun Li, Baosheng Li, Jian Zhu, Yong Yin, Zhenjiang Li
Purpose: Magnetic resonance (MR)-guided radiotherapy enables visualization of static anatomy, capturing tumor motion, and extracting quantitative image features for treatment verification and outcome monitoring. However, magnetic fields in online MR imaging (MRI) require efforts to ensure accurate dose measurements. This study aimed to assess the dosimetric impact of a 1.5 T magnetic field in esophageal cancer radiotherapy using MR-linac, exploring treatment adaptation potential and personalized medicine benefits. Methods: A prospective cohort study enrolled 100 esophageal squamous cell carcinoma patients undergoing 4DCT and 3DCT scans before radiotherapy. The heart was contoured on 3DCT, 4DCT end expiration (EE), and 4DCT end inhalation (EI) images by the same radiation oncologist. Reference RT plans were designed on 3DCT, with adjustments for different phases generating 5 plan types per patient. Variations in dose-volume parameters for organs at risk and the target area among different plans were compared using Monaco 5.40.04. Results: Slight dose distortions at air-tissue interfaces were observed in the magnetic field's presence. Dose at air-tissue interfaces (chest wall and heart wall) was slightly higher in some patients (3.0% tissue increased by 4.3 Gy on average) compared to nonmagnetic conditions. Average clinical target volume coverage V100 dropped from 99% to 95% compared to reference plans (planEI and planEE). Dose-volume histogram variation between the original plan and reference plans was within 2.3%. Superior-inferior (SI) direction displacement was significantly larger than lateral and anterior-posterior directions (P < .05). Conclusion: Significant SI direction shift in lower esophageal cancerous regions during RT indicates the magnetic field's dosimetric impact, including the electron return effect at tissue-air boundaries. Changes in OAR dose could serve as valuable indicators of organ impairment and target dose alterations, especially for cardiac tissue when using the 1.5 T linac method. Reoptimizing the plan with the magnetic field enhances the feasibility of achieving a clinically acceptable treatment plan for esophageal cancer patients.
{"title":"Assessing the Impact of a 1.5 T Transverse Magnetic Field in Radiotherapy for Esophageal Cancer Patients.","authors":"Yukun Li, Baosheng Li, Jian Zhu, Yong Yin, Zhenjiang Li","doi":"10.1177/15330338241227291","DOIUrl":"10.1177/15330338241227291","url":null,"abstract":"<p><p><b>Purpose:</b> Magnetic resonance (MR)-guided radiotherapy enables visualization of static anatomy, capturing tumor motion, and extracting quantitative image features for treatment verification and outcome monitoring. However, magnetic fields in online MR imaging (MRI) require efforts to ensure accurate dose measurements. This study aimed to assess the dosimetric impact of a 1.5 T magnetic field in esophageal cancer radiotherapy using MR-linac, exploring treatment adaptation potential and personalized medicine benefits. <b>Methods:</b> A prospective cohort study enrolled 100 esophageal squamous cell carcinoma patients undergoing 4DCT and 3DCT scans before radiotherapy. The heart was contoured on 3DCT, 4DCT end expiration (EE), and 4DCT end inhalation (EI) images by the same radiation oncologist. Reference RT plans were designed on 3DCT, with adjustments for different phases generating 5 plan types per patient. Variations in dose-volume parameters for organs at risk and the target area among different plans were compared using Monaco 5.40.04. <b>Results:</b> Slight dose distortions at air-tissue interfaces were observed in the magnetic field's presence. Dose at air-tissue interfaces (chest wall and heart wall) was slightly higher in some patients (3.0% tissue increased by 4.3 Gy on average) compared to nonmagnetic conditions. Average clinical target volume coverage V100 dropped from 99% to 95% compared to reference plans (planEI and planEE). Dose-volume histogram variation between the original plan and reference plans was within 2.3%. Superior-inferior (SI) direction displacement was significantly larger than lateral and anterior-posterior directions (<i>P</i> < .05). <b>Conclusion:</b> Significant SI direction shift in lower esophageal cancerous regions during RT indicates the magnetic field's dosimetric impact, including the electron return effect at tissue-air boundaries. Changes in OAR dose could serve as valuable indicators of organ impairment and target dose alterations, especially for cardiac tissue when using the 1.5 T linac method. Reoptimizing the plan with the magnetic field enhances the feasibility of achieving a clinically acceptable treatment plan for esophageal cancer patients.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241227291"},"PeriodicalIF":2.7,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10807384/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139521825","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Recent Advances and Challenges in Stereotactic Body Radiotherapy.","authors":"Atsuto Katano, Masanari Minamitani, Shingo Ohira, Hideomi Yamashita","doi":"10.1177/15330338241229363","DOIUrl":"10.1177/15330338241229363","url":null,"abstract":"","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241229363"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10851756/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139698371","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241235769
Minghui Zhang, Yan Wang, Mutian Lv, Li Sang, Xuemei Wang, Zijun Yu, Ziyi Yang, Zhongqing Wang, Liang Sang
Objectives: The purpose of this research is to summarize the structure of radiomics-based knowledge and to explore potential trends and priorities by using bibliometric analysis. Methods: Select radiomics-related publications from 2012 to October 2022 from the Science Core Collection Web site. Use VOSviewer (version 1.6.18), CiteSpace (version 6.1.3), Tableau (version 2022), Microsoft Excel and Rstudio's free online platforms (http://bibliometric.com) for co-writing, co-citing, and co-occurrence analysis of countries, institutions, authors, references, and keywords in the field. The visual analysis is also carried out on it. Results: The study included 6428 articles. Since 2012, there has been an increase in research papers based on radiomics. Judging by publications, China has made the largest contribution in this area. We identify the most productive institutions and authors as Fudan University and Tianjie. The top three magazines with the most publications are《FRONTIERS IN ONCOLOGY》, 《EUROPEAN RADIOLOGY》, and 《CANCERS》. According to the results of reference and keyword analysis, "deep learning, nomogram, ultrasound, f-18-fdg, machine learning, covid-19, radiogenomics" has been determined as the main research direction in the future. Conclusion: Radiomics is in a phase of vigorous development with broad prospects. Cross-border cooperation between countries and institutions should be strengthened in the future. It can be predicted that the development of deep learning-based models and multimodal fusion models will be the focus of future research. Advances in knowledge: This study explores the current state of research and hot spots in the field of radiomics from multiple perspectives, comprehensively, and objectively reflecting the evolving trends in imaging-related research and providing a reference for future research.
{"title":"Trends and Hotspots in Global Radiomics Research: A Bibliometric Analysis.","authors":"Minghui Zhang, Yan Wang, Mutian Lv, Li Sang, Xuemei Wang, Zijun Yu, Ziyi Yang, Zhongqing Wang, Liang Sang","doi":"10.1177/15330338241235769","DOIUrl":"10.1177/15330338241235769","url":null,"abstract":"<p><p><b>Objectives:</b> The purpose of this research is to summarize the structure of radiomics-based knowledge and to explore potential trends and priorities by using bibliometric analysis. <b>Methods:</b> Select radiomics-related publications from 2012 to October 2022 from the Science Core Collection Web site. Use VOSviewer (version 1.6.18), CiteSpace (version 6.1.3), Tableau (version 2022), Microsoft Excel and Rstudio's free online platforms (http://bibliometric.com) for co-writing, co-citing, and co-occurrence analysis of countries, institutions, authors, references, and keywords in the field. The visual analysis is also carried out on it. <b>Results:</b> The study included 6428 articles. Since 2012, there has been an increase in research papers based on radiomics. Judging by publications, China has made the largest contribution in this area. We identify the most productive institutions and authors as Fudan University and Tianjie. The top three magazines with the most publications are《FRONTIERS IN ONCOLOGY》, 《EUROPEAN RADIOLOGY》, and 《CANCERS》. According to the results of reference and keyword analysis, \"deep learning, nomogram, ultrasound, f-18-fdg, machine learning, covid-19, radiogenomics\" has been determined as the main research direction in the future. <b>Conclusion:</b> Radiomics is in a phase of vigorous development with broad prospects. Cross-border cooperation between countries and institutions should be strengthened in the future. It can be predicted that the development of deep learning-based models and multimodal fusion models will be the focus of future research. <b>Advances in knowledge:</b> This study explores the current state of research and hot spots in the field of radiomics from multiple perspectives, comprehensively, and objectively reflecting the evolving trends in imaging-related research and providing a reference for future research.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241235769"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10929060/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140094621","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241241245
Muhammet Ocak, Duygu Deniz Usta, Gokce Nur Arik Erol, Gulnur Take Kaplanoglu, Ece Konac, Atiye Seda Yar Saglam
Background: One of the most significant characteristics of cancer is epithelial-mesenchymal transition and research on the relationship between phenolic compounds and anticancer medications and epithelial-mesenchymal transition is widespread. Methods: In order to investigate the potential effects of Taxifolin on enhancing the effectiveness of Epirubicin in treating breast cancer, specifically in 4T1 cells and an allograft BALB/c model, the effects of Taxifolin and Epirubicin, both individually and in combination, were examined. Cell viability assays and cytotoxicity assays in 4T1 cells were performed. In addition, 4T1 cells were implanted into female BALB/c mice to conduct in vivo studies and evaluate the therapeutic efficacy of Taxifolin and Epirubicin alone or in combination. Tumor volumes and histological analysis were also assessed in mice. To further understand the mechanisms involved, we examined the messenger RNA and protein levels of epithelial-mesenchymal transition-related genes, as well as active Caspase-3/7 levels, using quantitative real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays, respectively. Results:In vitro results demonstrated that the coadministration of Taxifolin and Epirubicin reduced cell viability and cytotoxicity in 4T1 cell lines. In vivo, coadministration of Taxifolin and Epirubicin suppressed tumor growth in BALB/c mice with 4T1 breast cancer cells. Additionally, this combination treatment significantly increased the levels of active caspase-3/7 and downregulated the messenger RNA and protein levels of N-cadherin, β-catenin, vimentin, snail, and slug, but upregulated the E-cadherin gene. It significantly decreased the messenger RNA levels of the Zeb1 and Zeb2 genes. Conclusion: The in vitro and in vivo results of our study indicate that the concurrent use of Epirubicin with Taxifolin has supportive effects on breast cancer treatment.
{"title":"Determination of <i>In Vitro</i> and <i>In Vivo</i> Effects of Taxifolin and Epirubicin on Epithelial-Mesenchymal Transition in Mouse Breast Cancer Cells.","authors":"Muhammet Ocak, Duygu Deniz Usta, Gokce Nur Arik Erol, Gulnur Take Kaplanoglu, Ece Konac, Atiye Seda Yar Saglam","doi":"10.1177/15330338241241245","DOIUrl":"10.1177/15330338241241245","url":null,"abstract":"<p><p><b>Background:</b> One of the most significant characteristics of cancer is epithelial-mesenchymal transition and research on the relationship between phenolic compounds and anticancer medications and epithelial-mesenchymal transition is widespread. <b>Methods:</b> In order to investigate the potential effects of Taxifolin on enhancing the effectiveness of Epirubicin in treating breast cancer, specifically in 4T1 cells and an allograft BALB/c model, the effects of Taxifolin and Epirubicin, both individually and in combination, were examined. Cell viability assays and cytotoxicity assays in 4T1 cells were performed. In addition, 4T1 cells were implanted into female BALB/c mice to conduct <i>in vivo</i> studies and evaluate the therapeutic efficacy of Taxifolin and Epirubicin alone or in combination. Tumor volumes and histological analysis were also assessed in mice. To further understand the mechanisms involved, we examined the messenger RNA and protein levels of epithelial-mesenchymal transition-related genes, as well as active Caspase-3/7 levels, using quantitative real-time polymerase chain reaction, western blot, and enzyme-linked immunosorbent assays, respectively. <b>Results:</b> <i>In vitro</i> results demonstrated that the coadministration of Taxifolin and Epirubicin reduced cell viability and cytotoxicity in 4T1 cell lines. <i>In vivo</i>, coadministration of Taxifolin and Epirubicin suppressed tumor growth in BALB/c mice with 4T1 breast cancer cells. Additionally, this combination treatment significantly increased the levels of active caspase-3/7 and downregulated the messenger RNA and protein levels of N-cadherin, β-catenin, vimentin, snail, and slug, but upregulated the E-cadherin gene. It significantly decreased the messenger RNA levels of the Zeb1 and Zeb2 genes. <b>Conclusion:</b> The <i>in vitro</i> and <i>in vivo</i> results of our study indicate that the concurrent use of Epirubicin with Taxifolin has supportive effects on breast cancer treatment.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241241245"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10958820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140185596","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Introduction: We aimed to modify the LR-5 strategy to improve the diagnostic sensitivity for hepatocellular carcinoma (HCC) in high-risk patients while maintaining specificity. Methods: This study retrospectively analyzed 412 patients with 445 liver observations who underwent preoperative gadolinium ethoxybenzyl DTPA (GD-EOB-DTPA)-enhanced MRI followed by surgical procedures or biopsies. All observations were classified according to LI-RADS v2018, and the classifications were adjusted by modifying major features (MF)(substituting threshold growth with a more HCC-specific ancillary features (AF): presence of blood products within the mass, arterial phase hyperenhancement (APHE) was interpreted with hypointensity on precontrast imaging- isointensity in arterial phase (AP) and extending washout to transitional phase (TP)(2 min)). The specificity, sensitivity, and positive predictive value (PPV) were assessed to compare LR-5 (definitely HCC) diagnostic efficacy between LI-RADS version 2018 and modified LI-RADS. Results: Apart from nonenhancing "capsule", the interreader agreement of MFs and HCC-specific AFs between the two readers reached substantial or excellent ranges (κ values ranging from 0.631 to 0.911). According to LI-5 v2018, the specificity, sensitivity and PPV of HCC were 90.74%, 82.35%, and 98.17%, respectively. Based on a more HCC-specific AF, signal intensity in AP and TP (2 min), the sensitivity of the three modified strategies were 86.19%, 93.09%, 96.67% (P < .05)), while maintaining high specificity and PPV rates at 88.89% and 98.25% (P > .05) Conclusion: Further investigation into the efficacy of threshold growth as a MF is warranted. By utilizing GD-EOB-DTPA-enhanced MRI, enhancing the sensitivity of the modified LR-5 category may be achieved without compromising specificity and PPV in diagnosing HCC among high-risk patients.
{"title":"Based on Gadolinium Ethoxybenzyl DTPA-Enhanced MRI: Diagnostic Performance of the Category-Modified LR-5 Criteria in Patients At Risk for Hepatocellular Carcinoma.","authors":"Shaopeng Li, Kexue Deng, Jun Qiu, Peng Wang, Dawei Yin, Yiju Xie, Yongqiang Yu","doi":"10.1177/15330338241256859","DOIUrl":"10.1177/15330338241256859","url":null,"abstract":"<p><p><b>Introduction:</b> We aimed to modify the LR-5 strategy to improve the diagnostic sensitivity for hepatocellular carcinoma (HCC) in high-risk patients while maintaining specificity. <b>Methods:</b> This study retrospectively analyzed 412 patients with 445 liver observations who underwent preoperative gadolinium ethoxybenzyl DTPA (GD-EOB-DTPA)-enhanced MRI followed by surgical procedures or biopsies. All observations were classified according to LI-RADS v2018, and the classifications were adjusted by modifying major features (MF)(substituting threshold growth with a more HCC-specific ancillary features (AF): presence of blood products within the mass, arterial phase hyperenhancement (APHE) was interpreted with hypointensity on precontrast imaging- isointensity in arterial phase (AP) and extending washout to transitional phase (TP)(2 min)). The specificity, sensitivity, and positive predictive value (PPV) were assessed to compare LR-5 (definitely HCC) diagnostic efficacy between LI-RADS version 2018 and modified LI-RADS. <b>Results:</b> Apart from nonenhancing \"capsule\", the interreader agreement of MFs and HCC-specific AFs between the two readers reached substantial or excellent ranges (κ values ranging from 0.631 to 0.911). According to LI-5 v2018, the specificity, sensitivity and PPV of HCC were 90.74%, 82.35%, and 98.17%, respectively. Based on a more HCC-specific AF, signal intensity in AP and TP (2 min), the sensitivity of the three modified strategies were 86.19%, 93.09%, 96.67% (P < .05)), while maintaining high specificity and PPV rates at 88.89% and 98.25% (P > .05) <b>Conclusion:</b> Further investigation into the efficacy of threshold growth as a MF is warranted. By utilizing GD-EOB-DTPA-enhanced MRI, enhancing the sensitivity of the modified LR-5 category may be achieved without compromising specificity and PPV in diagnosing HCC among high-risk patients.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241256859"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11131403/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141082523","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-01-01DOI: 10.1177/15330338241258415
Junjie Zhang, Ligang Hao, Qian Xu, Fengxiao Gao
Objective: To develop and validate predictive models based on clinical parameters, and radiomic features to distinguish pulmonary pure invasive mucinous adenocarcinoma (pIMA) from mixed mucinous adenocarcinoma (mIMA) before surgery. Method: From January 2017 to December 2022, 193 pIMA and 111 mIMA were retrospectively analyzed at our hospital in this retrospective study. From contrast-enhanced computed tomography, 1037 radiomic features were extracted. The patients were randomly divided into a training group and a test group (n = 213 and 91, respectively) in a 7:3 ratio. The least absolute shrinkage and selection operator algorithm was used to select radiomic features. In this study, 9 machine learning radiomics prediction models were applied. The radiomics score was then calculated based on the best-performing machine learning model adopted. The clinical model was developed using the same machine learning model of radiomics. In the end, a combined model based on clinical factors and radiomics features was developed. The area under the receiver operating characteristic curve (AUC) value and decision curve analysis (DCA) were used to evaluate the clinical usefulness of the prediction model. Results: The combined model established by the Gaussian Naive Bayes machine learning method exhibited the best performance. The AUC of the combined model, clinical model, and radiomics model were 0.81, 0.80, and 0.68 in the training group and 0.91, 0.80, and 0.81 in the test group, respectively. The Brier scores of the combined model were 0.171 and 0.112. The DCA curve also showed that the combined model was beneficial to clinical settings. Conclusion: The combined model integration of radiomics features and clinical parameters may have potential value for the preoperative differentiation of pIMA from mIMA.
{"title":"Radiomics and Clinical Characters Based Gaussian Naive Bayes (GNB) Model for Preoperative Differentiation of Pulmonary Pure Invasive Mucinous Adenocarcinoma From Mixed Mucinous Adenocarcinoma.","authors":"Junjie Zhang, Ligang Hao, Qian Xu, Fengxiao Gao","doi":"10.1177/15330338241258415","DOIUrl":"10.1177/15330338241258415","url":null,"abstract":"<p><p><b>Objective:</b> To develop and validate predictive models based on clinical parameters, and radiomic features to distinguish pulmonary pure invasive mucinous adenocarcinoma (pIMA) from mixed mucinous adenocarcinoma (mIMA) before surgery. <b>Method:</b> From January 2017 to December 2022, 193 pIMA and 111 mIMA were retrospectively analyzed at our hospital in this retrospective study. From contrast-enhanced computed tomography, 1037 radiomic features were extracted. The patients were randomly divided into a training group and a test group (n = 213 and 91, respectively) in a 7:3 ratio. The least absolute shrinkage and selection operator algorithm was used to select radiomic features. In this study, 9 machine learning radiomics prediction models were applied. The radiomics score was then calculated based on the best-performing machine learning model adopted. The clinical model was developed using the same machine learning model of radiomics. In the end, a combined model based on clinical factors and radiomics features was developed. The area under the receiver operating characteristic curve (AUC) value and decision curve analysis (DCA) were used to evaluate the clinical usefulness of the prediction model. <b>Results:</b> The combined model established by the Gaussian Naive Bayes machine learning method exhibited the best performance. The AUC of the combined model, clinical model, and radiomics model were 0.81, 0.80, and 0.68 in the training group and 0.91, 0.80, and 0.81 in the test group, respectively. The Brier scores of the combined model were 0.171 and 0.112. The DCA curve also showed that the combined model was beneficial to clinical settings. <b>Conclusion:</b> The combined model integration of radiomics features and clinical parameters may have potential value for the preoperative differentiation of pIMA from mIMA.</p>","PeriodicalId":22203,"journal":{"name":"Technology in Cancer Research & Treatment","volume":"23 ","pages":"15330338241258415"},"PeriodicalIF":2.8,"publicationDate":"2024-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11143847/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141180662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}