1
|
Zheng J, Yan Z, Wang R, Xiao H, Chen Z, Ge X, Li Z, Liu Z, Yu H, Liu H, Wang G, Yu P, Fu J, Zhang G, Zhang J, Liu B, Huang Y, Deng H, Wang C, Fu W, Zhang Y, Wang R, Jiang Y, Lin Y, Huang L, Yang C, Cui F, He J, Liang H. NeoPred: dual-phase CT AI forecasts pathologic response to neoadjuvant chemo-immunotherapy in NSCLC. J Immunother Cancer 2025; 13:e011773. [PMID: 40449955 DOI: 10.1136/jitc-2025-011773] [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] [Accepted: 05/16/2025] [Indexed: 06/03/2025] Open
Abstract
BACKGROUND Accurate preoperative prediction of major pathological response or pathological complete response after neoadjuvant chemo-immunotherapy remains a critical unmet need in resectable non-small-cell lung cancer (NSCLC). Conventional size-based imaging criteria offer limited reliability, while biopsy confirmation is available only post-surgery. METHODS We retrospectively assembled 509 consecutive NSCLC cases from four Chinese thoracic-oncology centers (March 2018 to March 2023) and prospectively enrolled 50 additional patients. Three 3-dimensional convolutional neural networks (pre-treatment CT, pre-surgical CT, dual-phase CT) were developed; the best-performing dual-phase model (NeoPred) optionally integrated clinical variables. Model performance was measured by area under the receiver-operating-characteristic curve (AUC) and compared with nine board-certified radiologists. RESULTS In an external validation set (n=59), NeoPred achieved an AUC of 0.772 (95% CI: 0.650 to 0.895), sensitivity 0.591, specificity 0.733, and accuracy 0.627; incorporating clinical data increased the AUC to 0.787. In a prospective cohort (n=50), NeoPred reached an AUC of 0.760 (95% CI: 0.628 to 0.891), surpassing the experts' mean AUC of 0.720 (95% CI: 0.574 to 0.865). Model assistance raised the pooled expert AUC to 0.829 (95% CI: 0.707 to 0.951) and accuracy to 0.820. Marked performance persisted within radiological stable-disease subgroups (external AUC 0.742, 95% CI: 0.468 to 1.000; prospective AUC 0.833, 95% CI: 0.497 to 1.000). CONCLUSIONS Combining dual-phase CT and clinical variables, NeoPred reliably and non-invasively predicts pathological response to neoadjuvant chemo-immunotherapy in NSCLC, outperforms unaided expert assessment, and significantly enhances radiologist performance. Further multinational trials are needed to confirm generalizability and support surgical decision-making.
Collapse
Affiliation(s)
- Jianqi Zheng
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Zeping Yan
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Runchen Wang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Houlu Xiao
- Yuefa Health Technology (Guangzhou) Co., Ltd, Guangzhou, Guangdong, China
| | - Zhenlin Chen
- Yuefa Health Technology (Guangzhou) Co., Ltd, Guangzhou, Guangdong, China
| | - Xiaomin Ge
- Yuefa Health Technology (Guangzhou) Co., Ltd, Guangzhou, Guangdong, China
| | - Zhigang Li
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Zhichao Liu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Hong Yu
- Department of Radiology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, Shanghai, China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Guan Wang
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Pingwen Yu
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Junke Fu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jia Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Bohao Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Ying Huang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Hongshen Deng
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Chudong Wang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Wenhai Fu
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Yuan Zhang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Rui Wang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Yu Jiang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Yuechun Lin
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Linchong Huang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Chao Yang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Fei Cui
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| | - Hengrui Liang
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, Guangzhou, Guangdong, China
| |
Collapse
|
2
|
Chang YC, Nixon B, Souza F, Cardoso FN, Dayan E, Geiger EJ, Rosenberg A, D'Amato G, Subhawong T. The Desmoid Dilemma: Challenges and Opportunities in Assessing Tumor Burden and Therapeutic Response. Curr Oncol 2025; 32:288. [PMID: 40422547 DOI: 10.3390/curroncol32050288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/16/2025] [Accepted: 05/17/2025] [Indexed: 05/28/2025] Open
Abstract
Desmoid tumors are rare, locally invasive soft-tissue tumors with unpredictable clinical behavior. Imaging plays a crucial role in their diagnosis, measurement of disease burden, and assessment of treatment response. However, desmoid tumors' unique imaging features present challenges to conventional imaging metrics. The heterogeneous nature of these tumors, with a variable composition (fibrous, myxoid, or cellular), complicates accurate delineation of tumor boundaries and volumetric assessment. Furthermore, desmoid tumors can demonstrate prolonged stability or spontaneous regression, and biologic quiescence is often manifested by collagenization rather than bulk size reduction, making traditional size-based response criteria, such as Response Evaluation Criteria in Solid Tumors (RECIST), suboptimal. To overcome these limitations, advanced imaging techniques offer promising opportunities. Functional and parametric imaging methods, such as diffusion-weighted MRI, dynamic contrast-enhanced MRI, and T2 relaxometry, can provide insights into tumor cellularity and maturation. Radiomics and artificial intelligence approaches may enhance quantitative analysis by extracting and correlating complex imaging features with biological behavior. Moreover, imaging biomarkers could facilitate earlier detection of treatment efficacy or resistance, enabling tailored therapy. By integrating advanced imaging into clinical practice, it may be possible to refine the evaluation of disease burden and treatment response, ultimately improving the management and outcomes of patients with desmoid tumors.
Collapse
Affiliation(s)
- Yu-Cherng Chang
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bryan Nixon
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Felipe Souza
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fabiano Nassar Cardoso
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Etan Dayan
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Erik J Geiger
- Department of Orthopaedics, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Andrew Rosenberg
- Department of Pathology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Gina D'Amato
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ty Subhawong
- Department of Radiology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
3
|
Agnes A, Boldrini L, Perillo F, Tran HE, Brizi MG, Ricci R, Lenkowicz J, Votta C, Biondi A, Manfredi R, Valentini V, D'Ugo DM, Persiani R. Radiomic-based models are able to predict the pathologic response to different neoadjuvant chemotherapy regimens in patients with gastric and gastroesophageal cancer: a cohort study. World J Surg Oncol 2025; 23:183. [PMID: 40350424 PMCID: PMC12067740 DOI: 10.1186/s12957-025-03828-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/25/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND There is a clinical need to identify early predictors for response to neoadjuvant chemotherapy (NAC) in patients with gastric and gastroesophageal junction cancer (GC and GEJC). Radiomics involves extracting quantitative features from medical images. This study aimed to apply radiomics to build prediction models for the response to NAC. METHODS All consecutive patients with non-metastatic GC and GEJC undergoing NAC and surgical resection in an Italian high-volume referral center between 2005 and 2021 were considered eligible. In patients selected, the CT scans performed upon staging were reviewed to segment the tumor and extract radiomic features using MODDICOM. The primary endpoint was to develop and validate radiomic-based predictive models to identify major responders (MR: tumor regression grade TRG 1-2) and non-responders (NR: TRG 4-5) to NAC. Following an initial feature selection, radiomic and combined radiomic-clinicopathologic prediction models were built for the MR or NR status based on logistic regressions. Internal validation was performed for each model. Radiomic models (in the entire case series and according to NAC regimens) were evaluated using the receiver operating characteristic area under the curve (AUC), sensitivity, and negative predictive value (NPV). RESULTS The study included 77 patients undergoing NAC and subsequent tumor resection. The MR prediction model after all types of NAC (AUC of 0.876, CI 95% 0.786 - 0.966, sensitivity 83%, and NPV 96%) was based on a statistical feature. The models predicting NR among patients undergoing epirubicin with cisplatin and fluorouracil (ECF), epirubicin with oxaliplatin and capecitabin (EOX), or fluorouracil with oxaliplatin and docetaxel (FLOT) (AUC 0.760, CI 95% 0.639-0.882), oxaliplatin-based chemotherapy (AUC 0.810, CI 95% 0.692-0.928), and FLOT (AUC 0.907, CI 95% 0.818 - 0.995) were based on statistical, morphological and textural features. CONCLUSIONS The developed radiomic models resulted promising in predicting the response to different neoadjuvant chemotherapy strategies. Once further implemented on larger datasets, they could be valuable and cost-effective instruments to target multimodal treatment in patients with GC.
Collapse
Affiliation(s)
- Annamaria Agnes
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Luca Boldrini
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Federica Perillo
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
| | - Huong Elena Tran
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Maria Gabriella Brizi
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Riccardo Ricci
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Women, Children and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Jacopo Lenkowicz
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Claudio Votta
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Alberto Biondi
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy.
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy.
| | - Riccardo Manfredi
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Vincenzo Valentini
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Diagnostic Imaging, Oncological Radiotherapy and Hematology, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Domenico M D'Ugo
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| | - Roberto Persiani
- Catholic University of the Sacred Heart, Largo F. Vito n.1, Rome, 00168, Italy
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Largo A. Gemelli n. 8, Rome, 00168, Italy
| |
Collapse
|
4
|
Pourvaziri A, Mroueh N, Cochran RL, Srinivas Rao S, Kambadakone A. Beyond Conventional CT: Role of Dual-Energy CT in Monitoring Response to Therapy in Abdominal Malignancies. Radiol Imaging Cancer 2025; 7:e240142. [PMID: 40249270 DOI: 10.1148/rycan.240142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
In the era of precision medicine, imaging plays a critical role in evaluating treatment response to various oncologic therapies. For decades, conventional morphologic assessments using cross-sectional imaging have been the standard for monitoring the effectiveness of systemic and locoregional therapies in patients with cancer. However, the development of new functional imaging tools has widened the scope of imaging from mere response assessment to patient selection and outcome prediction. Dual-energy CT (DECT), known for its superior material differentiation capabilities, shows promise in enhancing treatment response evaluation. DECT-based iodine quantification methods are increasingly being investigated as surrogates for assessing tumor vascularity and physiology, which is particularly important in patients undergoing emerging targeted therapies. The purpose of this review article is to discuss the current and emerging role of DECT in assessing treatment response in patients with malignant abdominal tumors. Keywords: CT-Dual Energy, Transcatheter Tumor Therapy, Tumor Response, Iodine Uptake, Therapeutic Response © RSNA, 2025.
Collapse
Affiliation(s)
- Ali Pourvaziri
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Nayla Mroueh
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Rory L Cochran
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Shravya Srinivas Rao
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| |
Collapse
|
5
|
Spaanderman DJ, Starmans MPA, van Erp GCM, Hanff DF, Sluijter JH, Schut ARW, van Leenders GJLH, Verhoef C, Grünhagen DJ, Niessen WJ, Visser JJ, Klein S. Minimally interactive segmentation of soft-tissue tumors on CT and MRI using deep learning. Eur Radiol 2025; 35:2736-2745. [PMID: 39560714 PMCID: PMC12021718 DOI: 10.1007/s00330-024-11167-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/13/2024] [Accepted: 10/05/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Segmentations are crucial in medical imaging for morphological, volumetric, and radiomics biomarkers. Manual segmentation is accurate but not feasible in clinical workflow, while automatic segmentation generally performs sub-par. PURPOSE To develop a minimally interactive deep learning-based segmentation method for soft-tissue tumors (STTs) on CT and MRI. MATERIAL AND METHODS The interactive method requires the user to click six points near the tumor's extreme boundaries in the image. These six points are transformed into a distance map and serve, with the image, as input for a convolutional neural network. A multi-center public dataset with 514 patients and nine STT phenotypes in seven anatomical locations, with CT or T1-weighted MRI, was used for training and internal validation. For external validation, another public dataset was employed, which included five unseen STT phenotypes in extremities on CT, T1-weighted MRI, and T2-weighted fat-saturated (FS) MRI. RESULTS Internal validation resulted in a dice similarity coefficient (DSC) of 0.85 ± 0.11 (mean ± standard deviation) for CT and 0.84 ± 0.12 for T1-weighted MRI. External validation resulted in DSCs of 0.81 ± 0.08 for CT, 0.84 ± 0.09 for T1-weighted MRI, and 0.88 ± 0.08 for T2-weighted FS MRI. Volumetric measurements showed consistent replication with low error internally (volume: 1 ± 28 mm3, r = 0.99; diameter: - 6 ± 14 mm, r = 0.90) and externally (volume: - 7 ± 23 mm3, r = 0.96; diameter: - 3 ± 6 mm, r = 0.99). Interactive segmentation time was considerably shorter (CT: 364 s, T1-weighted MRI: 258s) than manual segmentation (CT: 1639s, T1-weighted MRI: 1895s). CONCLUSION The minimally interactive segmentation method effectively segments STT phenotypes on CT and MRI, with robust generalization to unseen phenotypes and imaging modalities. KEY POINTS Question Can this deep learning-based method segment soft-tissue tumors faster than can be done manually and more accurately than other automatic methods? Findings The minimally interactive segmentation method achieved accurate segmentation results in internal and external validation, and generalized well across soft-tissue tumor phenotypes and imaging modalities. Clinical relevance This minimally interactive deep learning-based segmentation method could reduce the burden of manual segmentation, facilitate the integration of imaging-based biomarkers (e.g., radiomics) into clinical practice, and provide a fast, semi-automatic solution for volume and diameter measurements (e.g., RECIST).
Collapse
Affiliation(s)
- Douwe J Spaanderman
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| | - Martijn P A Starmans
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Gonnie C M van Erp
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - David F Hanff
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Judith H Sluijter
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Anne-Rose W Schut
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Cornelis Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Wiro J Niessen
- Faculty of Medical Sciences, University of Groningen, Groningen, The Netherlands
| | - Jacob J Visser
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Stefan Klein
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Dioguardi Burgio M, Ronot M, Vilgrain V. ESR Essentials: assessing the radiological response of liver metastases to systemic therapy-practice recommendations by the European Society of Gastrointestinal and Abdominal Radiology. Eur Radiol 2025:10.1007/s00330-025-11540-1. [PMID: 40185923 DOI: 10.1007/s00330-025-11540-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 04/07/2025]
Abstract
The liver is a common site for metastatic spread, especially in advanced colorectal, breast, and pancreatic cancers. Imaging evaluation of liver metastases after systemic treatments like chemotherapy, targeted therapy, or immunotherapy is essential to distinguish treatment response from disease progression. The widely used response evaluation criteria in solid tumours (RECIST 1.1) focus on lesion size changes to evaluate treatment response. However, newer therapies, mainly targeted therapy and immunotherapy, often induce changes beyond size reduction, such as tumour necrosis, fibrosis, cystic transformation, calcifications, and modifications at the liver-tumour interface. These morphological and enhancement changes can be evaluated on CT and MRI and may better reflect the biological response in specific clinical settings. Overall, RECIST 1.1 criteria are recommended for assessing the radiological response of liver metastases after systemic treatment. The use of alternative radiological criteria validated on CT (such as Chun or Choi criteria) is recommended in specific clinical settings (e.g. metastatic colorectal cancer or metastatic gastrointestinal stromal tumours). Additionally, CT and MR modifications that reflect fibrosis, necrosis, calcifications, and haemorrhage can serve as ancillary indicators of tumoural response. These alternative criteria and radiological findings should be systematically assessed, particularly in liver metastases with minimal size changes, to better identify responders. KEY POINTS: RECIST 1.1 is the standard for evaluating tumour response in solid tumours and is recommended for the assessment of liver metastases after systemic therapy. CT attenuation, enhancement, and liver/tumour interface may correlate better with tumoural response compared to size reduction. CT and MR changes suggesting necrosis, fibrosis, calcifications, and haemorrhage can be used as additional indicators of tumoural response.
Collapse
Affiliation(s)
- Marco Dioguardi Burgio
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France.
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France.
| | - Maxime Ronot
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
| | - Valérie Vilgrain
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
| |
Collapse
|
7
|
Luo H, Gou YQ, Wang YS, Qin HL, Zhou HY, Zhang XM, Chen TW. A novel pN stage prediction model for resectable rectal adenocarcinoma based on preoperative MRI features and multiregional apparent diffusion coefficients. Eur Radiol 2025:10.1007/s00330-025-11528-x. [PMID: 40133439 DOI: 10.1007/s00330-025-11528-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/29/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025]
Abstract
OBJECTIVE To develop and validate a novel model based on preoperative MRI features and multiregional apparent diffusion coefficients (ADCs) to improve the prediction of pN stage in resectable rectal adenocarcinoma (RA). METHODS Two hundred fifty-four consecutive patients (median age [interquartile range], 67 [56-74] years; 156 males) with resectable RA were retrospectively collected at two medical centers from January 2017 to December 2023 and were divided into the training (n = 139), internal validation (n = 60), and external validation (n = 55) sets. All patients underwent preoperative MRI scans. Univariate and multivariate logistic regression analyses were conducted on the MRI features and multiregional (RA, peritumoral tissue, and tumor-adjacent rectal wall) ADCs to construct a nomogram model for preoperative predicting pN stage in the training set. Receiver operating characteristic (ROC) analysis was used to evaluate the predictive performance of the nomogram model vs the conventional MRI-assessed N (mriN) stage. The ROC curves were compared using the DeLong test. RESULTS The predictors incorporated in the nomogram model comprised gross tumor volume, categories of short diameter of maximum node, extramural vascular invasion, mesorectal fascia involvement, and ADCs of RA and peritumoral tissue. This model yielded a better prediction of the pN stage compared to the mriN stage in training (AUC, 0.848 vs 0.672; p < 0.001), internal validation (AUC, 0.843 vs 0.699; p = 0.008), and external validation (AUC, 0.857 vs 0.723; p = 0.01) sets. CONCLUSION This novel model based on the preoperative MRI features and multiregional ADCs can improve the prediction of the pN stage in RA. KEY POINTS Question Accurate preoperative assessment of the pN stage is important for determining an appropriate therapeutic strategy in rectal cancer, but the conventional mriN stage has low sensitivity. Findings Utilization of certain MRI features and multiregional ADCs improves preoperative assessment of the pN stage in RA when compared with conventional MRI assessment. Clinical relevance The novel model, based on preoperative MRI features and multiregional ADC values, can improve the prediction of the pN stage compared to the mriN stage in RA. The combination of this model with the mriN stage helps personalize treatment plans to improve patient prognosis.
Collapse
Affiliation(s)
- Hui Luo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yue-Qin Gou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yue-Su Wang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hui-Lin Qin
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hai-Ying Zhou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tian-Wu Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College, Nanchong, China.
| |
Collapse
|
8
|
Guha S, Ibrahim A, Geng P, Wu Q, Chou Y, Akin O, Schwartz LH, Xie CM, Zhao B. Variability of HCC Tumor Diameter and Density Measurements on Dynamic Contrast-Enhanced Computed Tomography. Tomography 2025; 11:36. [PMID: 40137576 PMCID: PMC11946049 DOI: 10.3390/tomography11030036] [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: 01/23/2025] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
PURPOSE In cancers imaged using contrast-enhanced protocols, such as hepatocellular carcinoma (HCC), formal guidelines rely on measurements of lesion size (in mm) and radiographic density (in Hounsfield units [HU]) to evaluate response to treatment. However, the variability of these measurements across different contrast enhancement phases remains poorly understood. This limits the ability of clinicians to discern whether measurement changes are accurate. METHODS In this study, we investigated the variability of maximal lesion diameter and mean lesion density of HCC lesions on CT scans across four different contrast enhancement phases: non-contrast-enhanced phase (NCE), early arterial phase (E-AP), late arterial phase (L-AP), and portal venous phase (PVP). HCC lesions were independently segmented by two expert radiologists. For each pair of a lesion's scan timepoints, one was selected randomly as the baseline measurement and the other as the repeat measurement. Both absolute and relative differences in measurements were calculated, as were the coefficients of variance (CVs). Analysis was further stratified by both contrast enhancement phase and lesion diameter. RESULTS Lesion diameter was found to have a CV of 5.11% (95% CI: 4.20-6.01%). About a fifth of the measurement's relative changes were greater than 10%. Although there was no significant difference in diameter measurements across different phases, there was a significant negative correlation (R = -0.303, p-value = 0.030) between lesion diameter and percent difference in diameter measurement. Lesion density measurements varied significantly across all phases, with the greatest relative difference of 47% in the late arterial phase and a CV of 22.84% (21.48-24.20%). The overall CV for lesion density measurements was 26.19% (24.66-27.72%). CONCLUSIONS Changes in tumor diameter measurements within 10% may simply be due to variability, and lesion density is highly sensitive to contrast timing. This highlights the importance of paying attention to these two variables when evaluating tumor response in both clinical trials and practice.
Collapse
Affiliation(s)
- Siddharth Guha
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (S.G.); (Y.C.)
| | - Abdalla Ibrahim
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| | - Pengfei Geng
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| | - Qian Wu
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| | - Yen Chou
- Department of Radiology, Columbia University Irving Medical Center, New York, NY 10032, USA; (S.G.); (Y.C.)
| | - Oguz Akin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| | - Lawrence H. Schwartz
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| | - Chuan-Miao Xie
- Sun Yat-sen University Cancer Center, Guangzhou 510060, China;
| | - Binsheng Zhao
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.I.); (P.G.); (Q.W.); (O.A.); (L.H.S.)
| |
Collapse
|
9
|
Zhao J, Bera K, Mohamed A, Li Q, Ramaiya N, Tirumani SH. Comparison of RECIST 1.1, mRECIST and PERCIST for assessment of peptide receptor radionuclide therapy treatment response in metastatic neuroendocrine tumors. Curr Probl Diagn Radiol 2025; 54:228-232. [PMID: 39389807 DOI: 10.1067/j.cpradiol.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 10/02/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE To compare RECIST 1.1, modified RECIST (mRECIST) and PERCIST for assessment of Peptide Receptor Radionuclide Therapy (PRRT) treatment response in metastatic neuroendocrine tumors. MATERIALS In this IRB-approved, HIPAA compliant retrospective study, patients treated with PRRT between July 2019 and Dec 2022 were identified. Inclusion criteria were presence of at least one pre-and one post-treatment imaging (CT, MRI, Ga 68 or Cu64 DOTATATE PET/CT) within one year of the start and end of PRRT respectively. The imaging was reviewed independently by two radiologists using RECIST 1.1, modified RECIST (mRECIST) and PERCIST criteria. Response of first post treatment scan and presence of disease progression during follow-up were recorded along with the date of best response and disease progression. Statistical analysis was performed to determine inter-reader agreement and agreement between the various response criteria using kappa statistics. RESULTS Best response by RECIST 1.1 was recorded in 26 patients (PR-7, SD- 13, PD- 6), by mRECIST in 22 patients (PR-7, SD- 10, PD- 5), by PERCIST in 14 patients (PR-4, SD- 3, PD- 7). Inter-reader agreement was highest for PERCIST (weighted kappa 0.921, standard error 0.078 95% CI 0.769 to 1.000) followed by RECIST 1.1 (weighted kappa 0.897, standard error 0.071 95% CI 0.758 to 1.000) and mRECIST (weighted kappa 0.883, standard error 0.079 95% CI 0.727 to 1.000).
Collapse
Affiliation(s)
- Jack Zhao
- Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Kaustav Bera
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States.
| | - Amr Mohamed
- Medical Oncology, Department of Hematology and Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Qiubai Li
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Nikhil Ramaiya
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Sree Harsha Tirumani
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
10
|
Chen S, Dai J, Zhao J, Han S, Zhang X, Chang J, Jiang D, Zhang H, Wang P, Hu S. Synthetic MRI Combined With Clinicopathological Characteristics for Pretreatment Prediction of Chemoradiotherapy Response in Advanced Nasopharyngeal Carcinoma. Korean J Radiol 2025; 26:135-145. [PMID: 39898394 PMCID: PMC11794295 DOI: 10.3348/kjr.2024.0385] [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: 10/31/2023] [Revised: 10/18/2024] [Accepted: 12/05/2024] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVE To explore the feasibility of synthetic magnetic resonance imaging (syMRI) combined with clinicopathological characteristics for the pre-treatment prediction of chemoradiotherapy (CRT) response in advanced nasopharyngeal carcinoma (ANPC). MATERIALS AND METHODS Patients with ANPC treated with CRT between September 2020 and June 2022 were retrospectively enrolled and categorized into response group (RG, n = 95) and non RGs (NRG, n = 32) based on the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1. The quantitative parameters from pre-treatment syMRI (longitudinal [T1] and transverse [T2] relaxation times and proton density [PD]), diffusion-weighted imaging (apparent diffusion coefficient [ADC]), and clinicopathological characteristics were compared between RG and NRG. Logistic regression analysis was applied to identify parameters independently associated with CRT response and to construct a multivariable model. The areas under the receiver-operating characteristic curve (AUC) for various diagnostic approaches were compared using the DeLong test. RESULTS The T1, T2, and PD values in the NRG were significantly lower than those in the RG (all P < 0.05), whereas no significant difference was observed in the ADC values between these two groups. Clinicopathological characteristics (Epstein-Barr virus [EBV]-DNA level, lymph node extranodal extension, clinical stage, and Ki-67 expression) exhibited significant differences between the two groups. Logistic regression analysis showed that T1, PD, EBV-DNA level, clinical stage, and Ki-67 expression had significant independent relationships with CRT response (all P < 0.05). The multivariable model incorporating these five variables yielded AUC, sensitivity, and specificity values of 0.974, 93.8% (30/32), and 91.6% (87/95), respectively. CONCLUSION SyMRI may be used for the pretreatment prediction of CRT response in ANPC. The multivariable model incorporating syMRI quantitative parameters and clinicopathological characteristics, which were independently associated with CRT response, may be a new tool for the pretreatment prediction of CRT response.
Collapse
Affiliation(s)
- Siyu Chen
- Department of Intensive Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jiankun Dai
- GE Healthcare, MR Research China, Beijing, China
| | - Jing Zhao
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Shuang Han
- Department of Pathology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Xiaojun Zhang
- Department of Radiation Oncology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Jun Chang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Donghui Jiang
- Department of Intensive Medicine, Affiliated Hospital of Jiangnan University, Wuxi, China
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Heng Zhang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Peng Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China.
| | - Shudong Hu
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
11
|
Luo H, Gou YQ, Wang YS, Qin HL, Zhou HY, Zhang XM, Chen TW. Comparison of apparent diffusion coefficients of resectable mid‑high rectal adenocarcinoma and distal paracancerous tissue. Oncol Lett 2025; 29:97. [PMID: 39697979 PMCID: PMC11653244 DOI: 10.3892/ol.2024.14843] [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: 07/06/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Paracancerous tissues actively communicate with the tumor and undergo molecular alterations associated with tumorigenesis. Apparent diffusion coefficient (ADC) can help distinguish between rectal adenocarcinoma (RA), tumor-adjacent and tumor-distant tissues. Preoperative determining optimal distal resection margin (DRM) is crucial for formulating surgical options. The present study aimed to assess ADC differences between RA and 1 cm-layer distal paracancerous tissues, providing a potential reference basis for preoperatively determining optimal DRM. A total of 110 consecutive patients with mid-high RA undergoing preoperative diffusion-weighted imaging were included. ADCs of RA and distal paracancerous tissues located ~1, 2 and 3 cm from the tumor margin (defined as D1, D2 and D3, respectively) were measured using five b-value pairs (0 and 50; 0 and 100; 0 and 800; 0 and 1,000; and 0 and 1,500 sec/mm2). Differences in ADCs between RA, D1, D2 and D3 were compared using the Friedman test with a post hoc Bonferroni correction. Variables that demonstrated statistical differences in multiple pairwise comparisons underwent receiver operating characteristic (ROC) analysis to assess diagnostic performance of ADCs in distinguishing between tissues. ADC at all b-value pairs demonstrated satisfactory performance in distinguishing RA from D1, D2 and D3 [areas under the ROC curves (AUCs), 0.838 to 0.996)]. When the maximum b-value was ≥800 sec/mm2, the ADC of D1 was significantly lower compared with those of D2 and D3 (P<0.001). ADC exhibited an optimal performance in differentiating D1 from D2 at b-values of 0 and 800 sec/mm2, and D1 from D3 at b-values of 0 and 1,000 sec/mm2 (AUCs: 0.652 and 0.692, respectively). However, ADCs of D2 and D3 demonstrated no differences at all b-value pairs (all P>0.05). In conclusion, ADC may distinguish RA from D1, D2 and D3, and D1 from D2/D3, but cannot distinguish between D2 and D3.
Collapse
Affiliation(s)
- Hui Luo
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yue-Qin Gou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Yue-Su Wang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hui-Lin Qin
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Hai-Ying Zhou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Tian-Wu Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
12
|
Thater G, Frerichs I, Büttner S, Schoenberg SO, Froelich M, Ayx I. Reduction of Streak Artifacts in the Superior Vena Cava for Better Visualization of Mediastinal Structures Through Virtual Monoenergetic Reconstructions Using a Photon-counting Detector Computed Tomography. J Thorac Imaging 2025:00005382-990000000-00163. [PMID: 39885700 DOI: 10.1097/rti.0000000000000822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
PURPOSE Computed tomography (CT) is crucial in oncologic imaging for precise diagnosis and staging. Beam-hardening artifacts from contrast media in the superior vena cava can degrade image quality and obscure adjacent structures, complicating lymph node assessment. This study examines the use of virtual monoenergetic reconstruction with photon-counting detector CT (photon-counting CT) to mitigate these artifacts. MATERIALS AND METHODS The retrospective study included 50 patients who underwent thoracoabdominal scans. Virtual monoenergetic reconstructions at nine keV levels (60 to 140 keV) were analyzed for Hounsfield Unit (HU) stability, image noise, and artifact index in various regions of interest (ROIs): mediastinal adipose tissue (ROI 1 to 3) and vascular stations (ROI 4 to 6) were compared with reference tissue (ROI 7 to 8). The diagnostic image quality of the keV levels was assessed using a 5-point Likert Scale. RESULTS Lower keV values (60 to 80) exhibited higher image noise and lower HU stability in mediastinal adipose tissue compared with higher energies, with optimal noise reduction observed at 130 keV (ROI 1 to 3). HU stability in vascular structures (ROI 4 to 6) significantly improved above 80 keV, with the best performance at 140 keV. Artifact levels decreased progressively from 60 to 140 keV. Visually, keV levels of 110 keV (96% Likert ≥4) and 120 keV (60% Likert 4) were rated most diagnostically valuable, consistent with technical findings. CONCLUSION Virtual monoenergetic reconstructions with photon-counting CT effectively reduce beam-hardening artifacts near the superior vena cava, enhancing the visualization of lymph nodes and adjacent structures. This technology advances oncologic imaging by improving diagnostic accuracy in areas previously affected by artifact-related image degradation.
Collapse
Affiliation(s)
- Greta Thater
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim
| | - Isabel Frerichs
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim
| | - Sylvia Büttner
- Department of Medical Statistics, Biomathematics and Information Processing, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan O Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim
| | - Matthias Froelich
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim
| | - Isabelle Ayx
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim
| |
Collapse
|
13
|
Wang F, Wang Y, Xiong B, Yang Z, Wang J, Yao Y, Yu L, Fu Q, Li L, Zhang Q, Zheng C, Huang S, Liu L, Liu C, Sun H, Mao B, Liu DX, Yu Z. Neoadjuvant pyrotinib and trastuzumab in HER2-positive breast cancer with no early response (NeoPaTHer): efficacy, safety and biomarker analysis of a prospective, multicentre, response-adapted study. Signal Transduct Target Ther 2025; 10:45. [PMID: 39875376 PMCID: PMC11775149 DOI: 10.1038/s41392-025-02138-6] [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: 08/12/2024] [Revised: 12/31/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
The potential benefits of pyrotinib for patients with trastuzumab-insensitive, HER2-positive early-stage breast cancer remain unclear. This prospective, multicentre, response-adapted study evaluated the efficacy and safety of adding pyrotinib to the neoadjuvant treatment of HER2-positive breast cancer patients with a poor response to initial docetaxel plus carboplatin and trastuzumab (TCbH). Early response was assessed using magnetic resonance imaging (MRI) after two cycles of treatment. Patients showing poor response, as defined by RECIST 1.1, could opt to receive additional pyrotinib or continue standard therapy. The primary endpoint was the total pathological complete response (tpCR; ypT0/isN0) rate. Of the 129 patients enroled, 62 (48.1%) were identified as MRI-responders (cohort A), 26 non-responders continued with four more cycles of TCbH (cohort B), and 41 non-responders received additional pyrotinib (cohort C). The tpCR rate was 30.6% (95% CI: 20.6-43.0%) in cohort A, 15.4% (95% CI: 6.2-33.5%) in cohort B, and 29.3% (95% CI: 17.6-44.5%) in cohort C. Multivariable logistic regression analyses demonstrated comparable odds of achieving tpCR between cohorts A and C (odds ratio = 1.04, 95% CI: 0.40-2.70). No new adverse events were observed with the addition of pyrotinib. Patients with co-mutations of TP53 and PIK3CA exhibited lower rates of early partial response compared to those without or with a single gene mutation (36.0% vs. 60.0%, P = 0.08). These findings suggest that adding pyrotinib may benefit patients who do not respond to neoadjuvant trastuzumab plus chemotherapy. Further investigation is warranted to identify biomarkers predicting patients' benefit from the addition of pyrotinib.
Collapse
Affiliation(s)
- Fei Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Yongjiu Wang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Bin Xiong
- Department of Breast Surgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Zhenlin Yang
- Department of Breast Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jingfen Wang
- Department of Breast Surgery, Linyi Cancer Hospital, Linyi, China
| | - Yumin Yao
- Department of Breast Surgery, Liaocheng People's Hospital, Liaocheng, China
| | - Lixiang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Qinye Fu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Liang Li
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Qiang Zhang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
| | - Chao Zheng
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Shuya Huang
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Liyuan Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Chun Liu
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Huaibo Sun
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Beibei Mao
- Genecast Biotechnology Co.Ltd., Wuxi, China
| | - Dong-Xu Liu
- Breast Center, The Second Hospital of Shandong University, Jinan, China
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China
| | - Zhigang Yu
- Breast Center, The Second Hospital of Shandong University, Jinan, China.
- Shandong Key Laboratory of Cancer Digital Medicine, Jinan, China.
- Shandong Provincial Engineering Laboratory of Translational Research on Prevention and Treatment of Breast Disease, Jinan, China.
- Institute of Translational Medicine of Breast Disease Prevention and Treatment, Shandong University, Jinan, China.
| |
Collapse
|
14
|
Graf M, Ziegelmayer S, Reischl S, Teumer Y, Gassert FT, Marka AW, Raffler P, Bachmann J, Makowski M, Reim D, Lohöfer F, Burian E, Braren R. CT-Derived Quantitative Image Features Predict Neoadjuvant Treatment Response in Adenocarcinoma of the Gastroesophageal Junction with High Accuracy. Cancers (Basel) 2025; 17:216. [PMID: 39857998 PMCID: PMC11763438 DOI: 10.3390/cancers17020216] [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: 12/03/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The purpose of this retrospective study was to evaluate the value of contrast-enhanced computed tomography (CE-CT) image features at baseline and after neoadjuvant chemotherapy in predicting histopathological response in patients with adenocarcinoma of the gastroesophageal junction (GEJ). METHODS A total of 105 patients with a diagnosis of adenocarcinoma of the GEJ were examined by CE-CT at baseline and preoperatively after neoadjuvant chemotherapy. All patients underwent surgical resection. Histopathological parameters and tumor regression grading according to Becker et al. were collected in 93 patients. Line profiles of the primary tumor area in baseline and preoperative CE-CT were generated using ImageJ. Maximum tumor density and tumor-to-wall density delta were calculated and correlated with the histopathological tumor response. In addition, tumor response was assessed according to standard RECIST measurements in all patients and by endoscopy in 72 patients. RESULTS Baseline and change in baseline to preoperative CE-CT parameters showed no significant differences between responders (Becker grade 1a, 1b) and non-responders (Becker grade 2, 3). After neoadjuvant therapy, responders and non-responders showed significant differences in maximum density and tumor-to-wall density delta values. Line profile measurements showed excellent inter-rater agreement. In comparison, neither RECIST nor endoscopy showed significant differences between these groups. CONCLUSIONS Posttreatment CE-CT can predict histopathological therapy response to neoadjuvant treatment in adenocarcinoma of GEJ patients with high accuracy and thus may improve patient management.
Collapse
Affiliation(s)
- Markus Graf
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Sebastian Ziegelmayer
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Stefan Reischl
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Yannick Teumer
- Department of Medicine II, Ulm University Medical Center, 89081 Ulm, Germany;
| | - Florian T. Gassert
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Alexander W. Marka
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Philipp Raffler
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Jeannine Bachmann
- Department of Surgery, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (J.B.); (D.R.)
| | - Marcus Makowski
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Daniel Reim
- Department of Surgery, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (J.B.); (D.R.)
| | - Fabian Lohöfer
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
| | - Egon Burian
- Diagnostic and Interventional Radiology, University Hospital Zurich, University Zurich, 8006 Zurich, Switzerland;
- Faculty of Medicine, University of Zurich, 8006 Zurich, Switzerland
| | - Rickmer Braren
- Department of Diagnostic and Interventional Radiology, School of Medicine & Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany; (S.Z.); (S.R.); (F.T.G.); (A.W.M.); (P.R.); (M.M.); (F.L.); (R.B.)
- German Cancer Consortium (DKTK, Partner Site Munich), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
15
|
Zhang J, Wei L, Zhou F, Du Z, Wang M, Wu G, Yuan Q, Xi C, Yang W, Fu P, Wu B, Yu J, Hu J. Remodeling and Characterization Analysis of Corticospinal Tract in Patients with Intracerebral Hemorrhage in the Basal Ganglia. Transl Stroke Res 2025:10.1007/s12975-025-01326-4. [PMID: 39786647 DOI: 10.1007/s12975-025-01326-4] [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: 04/15/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
To investigate corticospinal tract (CST) injury and remodeling in patients with basal ganglia intracerebral hemorrhage (ICH) and explore the characterization capabilities of the corresponding parameters. In this prospective study, baseline, scale, and diffusion-weighted imaging (DWI) data were collected from patient cohorts. Participants were stratified into favorable (0-3 points) and unfavorable (4-6 points) prognosis groups, based on Modified Rankin Scale (mRS) after 3-6 months. The analysis of DWI data was conducted employing FSL and DSI Studio software to compare CST injury between the prognosis groups and CST remodeling features. A partial correlation model was deployed to elucidate the characterization capability of CST-related parameters. Additionally, logistic regression analysis was applied to identify factors significantly influencing prognosis. A total of 65 patients were enrolled with a mean age of 53.52 years and a median hematoma volume of 23.60 ml. The 44 patients were classified within the favorable prognosis group, demonstrating a statistically significant difference in their lower mean age (P = 0.002). Additionally, 10 patients underwent DWI review with a mean age of 50.30 years and a median hematoma volume of 18.56 ml. The investigation uncovered evidence of CST damage versus remodeling at the group level, respectively, with statistical significance (FDR-corrected P < 0.05, 10,000 permutations). The fractional anisotropy (FA) ratio in the internal capsule region exhibited moderate correlation with motor function (r = 0.507, P < 0.001) and the 3- to 6-month mRS scores (r = - 0.318, P < 0.013). Furthermore, binary logistic regression analysis identified the FA rate in the internal capsule as a significant influencing factor of prognosis (odds ratio = 1.027, 95% confidence interval = 1.003-1.052, P = 0.025). Basal ganglia ICH can coincide with injury to the CST, which could undergo repair over time. Additionally, the FA ratio of the internal capsule is a potential biomarker to characterize residual motor function and provide prognostic information.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Lichao Wei
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Fengyuan Zhou
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Zhuoyin Du
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Meihua Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Gang Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Qiang Yuan
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Caihua Xi
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Weijian Yang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Pengfei Fu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Biwu Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China
| | - Jian Yu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai, 200040, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China.
| | - Jin Hu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai, 200040, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200040, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, 200040, China.
| |
Collapse
|
16
|
Chen YH, Lue KH, Chu SC, Lin CB, Liu SH. The value of 18F-fluorodeoxyglucose positron emission tomography-based radiomics in non-small cell lung cancer. Tzu Chi Med J 2025; 37:17-27. [PMID: 39850392 PMCID: PMC11753514 DOI: 10.4103/tcmj.tcmj_124_24] [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: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/24/2024] [Indexed: 01/25/2025] Open
Abstract
Currently, the second most commonly diagnosed cancer in the world is lung cancer, and 85% of cases are non-small cell lung cancer (NSCLC). With growing knowledge of oncogene drivers and cancer immunology, several novel therapeutics have emerged to improve the prognostic outcomes of NSCLC. However, treatment outcomes remain diverse, and an accurate tool to achieve precision medicine is an unmet need. Radiomics, a method of extracting medical imaging features, is promising for precision medicine. Among all radiomic tools, 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET)-based radiomics provides distinct information on glycolytic activity and heterogeneity. In this review, we collected relevant literature from PubMed and summarized the various applications of 18F-FDG PET-derived radiomics in improving the detection of metastasis, subtyping histopathologies, characterizing driver mutations, assessing treatment response, and evaluating survival outcomes of NSCLC. Furthermore, we reviewed the values of 18F-FDG PET-based deep learning. Finally, several challenges and caveats exist in the implementation of 18F-FDG PET-based radiomics for NSCLC. Implementing 18F-FDG PET-based radiomics in clinical practice is necessary to ensure reproducibility. Moreover, basic studies elucidating the underlying biological significance of 18F-FDG PET-based radiomics are lacking. Current inadequacies hamper immediate clinical adoption; however, radiomic studies are progressively addressing these issues. 18F-FDG PET-based radiomics remains an invaluable and indispensable aspect of precision medicine for NSCLC.
Collapse
Affiliation(s)
- Yu-Hung Chen
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Kun-Han Lue
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sung-Chao Chu
- School of Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Hematology and Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Bin Lin
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shu-Hsin Liu
- Department of Medical Imaging and Radiological Sciences, Tzu Chi University, Hualien, Taiwan
- Department of Nuclear Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| |
Collapse
|
17
|
Liu H, Ibrahim EIK, Centanni M, Sarr C, Venkatakrishnan K, Friberg LE. Integrated modeling of biomarkers, survival and safety in clinical oncology drug development. Adv Drug Deliv Rev 2025; 216:115476. [PMID: 39577694 DOI: 10.1016/j.addr.2024.115476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/12/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Model-based approaches, including population pharmacokinetic-pharmacodynamic modeling, have become an essential component in the clinical phases of oncology drug development. Over the past two decades, models have evolved to describe the temporal dynamics of biomarkers and tumor size, treatment-related adverse events, and their links to survival. Integrated models, defined here as models that incorporate at least two pharmacodynamic/ outcome variables, are applied to answer drug development questions through simulations, e.g., to support the exploration of alternative dosing strategies and study designs in subgroups of patients or other tumor indications. It is expected that these pharmacometric approaches will be expanded as regulatory authorities place further emphasis on early and individualized dosage optimization and inclusive patient-focused development strategies. This review provides an overview of integrated models in the literature, examples of the considerations that need to be made when applying these advanced pharmacometric approaches, and an outlook on the expected further expansion of model-informed drug development of anticancer drugs.
Collapse
Affiliation(s)
- Han Liu
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Eman I K Ibrahim
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Maddalena Centanni
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden
| | - Céline Sarr
- Pharmetheus AB, Dragarbrunnsgatan 77, 753 19, Uppsala, Sweden
| | | | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Box 580, 75123, Uppsala, Sweden.
| |
Collapse
|
18
|
Jungblut L, Rizzo SM, Ebner L, Kobe A, Nguyen-Kim TDL, Martini K, Roos J, Puligheddu C, Afshar-Oromieh A, Christe A, Dorn P, Funke-Chambour M, Hötker A, Frauenfelder T. Advancements in lung cancer: a comprehensive perspective on diagnosis, staging, therapy and follow-up from the SAKK Working Group on Imaging in Diagnosis and Therapy Monitoring. Swiss Med Wkly 2024; 154:3843. [PMID: 39835913 DOI: 10.57187/s.3843] [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: 01/22/2025] Open
Abstract
In 2015, around 4400 individuals received a diagnosis of lung cancer, and Switzerland recorded approximately 3200 deaths related to lung cancer. Advances in detection, such as lung cancer screening and improved treatments, have led to increased identification of early-stage lung cancer and higher chances of long-term survival. This progress has introduced new considerations in imaging, emphasising non-invasive diagnosis and characterisation techniques like radiomics. Treatment aspects, such as preoperative assessment and the implementation of immune response evaluation criteria in solid tumours (iRECIST), have also seen advancements. For those undergoing curative treatment for lung cancer, guidelines propose follow-up with computed tomography (CT) scans within a specific timeframe. However, discrepancies exist in published guidelines, and there is a lack of universally accepted recommendations for follow-up procedures. This white paper aims to provide a certain standard regarding the use of imaging on the diagnosis, staging, treatment and follow-up of patients with lung cancer.
Collapse
Affiliation(s)
- Lisa Jungblut
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefania Maria Rizzo
- Service of Radiology, Imaging Institute of Southern Switzerland, Clinica Di Radiologia EOC, Lugano, Switzerland
| | - Lukas Ebner
- Department of Radiology and Nuclear Medicine, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Adrian Kobe
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thi Dan Linh Nguyen-Kim
- Institute of Radiology and Nuclear Medicine, Stadtspital Triemli Zurich, Zurich, Switzerland
| | - Katharina Martini
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Justus Roos
- Department of Radiology and Nuclear Medicine, Luzerner Kantonsspital, Lucerne, Switzerland
| | - Carla Puligheddu
- Imaging Institute of Southern Switzerland (IIMSI), Ente Ospedaliero Cantonale (EOC), Lugano, Switzerland
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andreas Christe
- Department of Radiology SLS, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andreas Hötker
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Thomas Frauenfelder
- Diagnostic and Interventional Radiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Sijens PE. Editorial for " 31P MR Spectroscopy in the Pancreas: Repeatability, Comparison With Liver, and Pilot Pancreatic Cancer Data". J Magn Reson Imaging 2024; 60:2667-2668. [PMID: 38485513 DOI: 10.1002/jmri.29330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 11/15/2024] Open
Affiliation(s)
- Paul E Sijens
- Department of Radiology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
20
|
Illy M, Bartoli A, Mancini J, Duffaud F, Vidal V, Tradi F. Dedicated software to harmonize the follow-up of oncological patients. RESEARCH IN DIAGNOSTIC AND INTERVENTIONAL IMAGING 2024; 12:100051. [PMID: 39391594 PMCID: PMC11462215 DOI: 10.1016/j.redii.2024.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 08/04/2024] [Indexed: 10/12/2024]
Abstract
Objective To test and evaluate a sofware dedicated to the follow-up of oncological CT scans for potential use in the Radiology department. Materials and methods In this retrospective study, 37 oncological patients with baseline and follow-up CT scans were reinterpreted using a dedicated software. Baseline CT scans were chosen from the imaging reports available in our PACS (picture archiving and communicatin systems). Follow-up interpretations were independently assessed with the software. We evaluated the target lesion sums and the tumor response based on RECIST 1.1 (Response Evaluation Criteria in Solid Tumors). Results There was no significant difference in the target lesion sums and the tumor response assessments between the PACS data and the imaging software. There was no over or underestimation of the disease with the software. There was a sigificant deviation (progression versus stability) in three cases. For two patients, this difference was related to the evaluation of the response of non-target lesions. The difference in the third patient was due to comparison with a previous CT scan than to the baseline exam. There was a miscalculation in 13 % of the reports and in 28 % of the cases the examination was compared to the previous CT scan. Finally, the tumor response was not detailed in 43 % of the follow-up reports. Conclusion The use of dedicated oncology monitoring software could help in reducing intepretation time and in limiting human errors.
Collapse
Affiliation(s)
- Mathias Illy
- Radiology Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Axel Bartoli
- Radiology Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Julien Mancini
- Public Health Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Florence Duffaud
- Oncology Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Vincent Vidal
- Radiology Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Farouk Tradi
- Radiology Department, hôpital de la Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| |
Collapse
|
21
|
Xiuli N, Hua C, Peng G, Hairong Y, Meili S, Peng Y. Feasibility of an artificial intelligence system for tumor response evaluation. BMC Med Imaging 2024; 24:280. [PMID: 39425045 PMCID: PMC11488245 DOI: 10.1186/s12880-024-01460-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
PURPOSE The objective of this study was to evaluate the feasibility of using Artificial Intelligence (AI) to measure the long-diameter of tumors for evaluating treatment response. METHODS Our study included 48 patients with lung-specific target lesions and conducted 277 measurements. The radiologists recorded the long-diameter in axial imaging plane of the target lesions for each measurement. Meanwhile, AI software was utilized to measure the long-diameter in both the axial imaging plane and in three dimensions (3D). Statistical analyses including the Bland-Altman plot, Spearman correlation analysis, and paired t-test to ascertain the accuracy and reliability of our findings. RESULTS The Bland-Altman plot showed that the AI measurements had a bias of -0.28 mm and had limits of agreement ranging from - 13.78 to 13.22 mm (P = 0.497), indicating agreement with the manual measurements. However, there was no agreement between the 3D measurements and the manual measurements, with P < 0.001. The paired t-test revealed no statistically significant difference between the manual measurements and AI measurements (P = 0.497), whereas a statistically significant difference was observed between the manual measurements and 3D measurements (P < 0.001). CONCLUSIONS The application of AI in measuring the long-diameter of tumors had significantly improved efficiency and reduced the incidence of subjective measurement errors. This advancement facilitated more convenient and accurate tumor response evaluation.
Collapse
Affiliation(s)
- Nie Xiuli
- Department of Radiology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Chen Hua
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Gao Peng
- Department of Radiology, Jiaozhou Hospital of Tongji University Dongfang Hospital, Tongji University, Qingdao, China
| | - Yu Hairong
- Department of Radiology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Sun Meili
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Yan Peng
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China.
| |
Collapse
|
22
|
Luo C, Huang W, Li S, Li H, Ruan G, Fu G, Liu L. Optimal cut-off value for identifying objective response in patients with nasopharyngeal carcinoma after induction chemotherapy. Head Neck 2024; 46:2540-2549. [PMID: 38545637 DOI: 10.1002/hed.27754] [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: 09/27/2023] [Revised: 01/20/2024] [Accepted: 03/14/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND We aimed to establish the most suitable threshold for objective response (OR) in the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 in patients with nasopharyngeal carcinoma (NPC). METHODS According to RECIST 1.1, we retrospectively evaluated MR images of NPC lesions in patients before and after induction chemotherapy (IC). Restricted cubic spline and maximally selected rank statistics were used to determine the cut-off value. Survival rates and differences between groups were compared with Kaplan-Meier curves and log-rank tests. RESULTS Of 1126 patients, 365 cases who received IC treatment were suitable for RECIST 1.1 evaluation. The 20% cut-off value maximized between-group differences according to maximally selected rank statistics. No difference in distant metastasis-free survival between OR and non-response groups was shown using the primary threshold of OR (30%), while it differed when 20% was employed. CONCLUSIONS With an optimal cut-off value of 20%, RECIST may assist clinicians to accurately evaluate disease response in NPC patients.
Collapse
Affiliation(s)
- Chao Luo
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Wenjie Huang
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuqi Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Haojiang Li
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Guangying Ruan
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gui Fu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Lizhi Liu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Radiology, The Third People's Hospital of Shenzhen, Shenzhen, China
| |
Collapse
|
23
|
Wang SX, Yang Y, Xie H, Yang X, Liu ZQ, Li HJ, Huang WJ, Luo WJ, Lei YM, Sun Y, Ma J, Chen YF, Liu LZ, Mao YP. Radiomics-based nomogram guides adaptive de-intensification in locoregionally advanced nasopharyngeal carcinoma following induction chemotherapy. Eur Radiol 2024; 34:6831-6842. [PMID: 38514481 DOI: 10.1007/s00330-024-10678-8] [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: 11/09/2023] [Revised: 01/13/2024] [Accepted: 02/07/2024] [Indexed: 03/23/2024]
Abstract
OBJECTIVES This study aimed to construct a radiomics-based model for prognosis and benefit prediction of concurrent chemoradiotherapy (CCRT) versus intensity-modulated radiotherapy (IMRT) in locoregionally advanced nasopharyngeal carcinoma (LANPC) following induction chemotherapy (IC). MATERIALS AND METHODS A cohort of 718 LANPC patients treated with IC + IMRT or IC + CCRT were retrospectively enrolled and assigned to a training set (n = 503) and a validation set (n = 215). Radiomic features were extracted from pre-IC and post-IC MRI. After feature selection, a delta-radiomics signature was built with LASSO-Cox regression. A nomogram incorporating independent clinical indicators and the delta-radiomics signature was then developed and evaluated for calibration and discrimination. Risk stratification by the nomogram was evaluated with Kaplan-Meier methods. RESULTS The delta-radiomics signature, which comprised 19 selected features, was independently associated with prognosis. The nomogram, composed of the delta-radiomics signature, age, T category, N category, treatment, and pre-treatment EBV DNA, showed great calibration and discrimination with an area under the receiver operator characteristic curve of 0.80 (95% CI 0.75-0.85) and 0.75 (95% CI 0.64-0.85) in the training and validation sets. Risk stratification by the nomogram, excluding the treatment factor, resulted in two groups with distinct overall survival. Significantly better outcomes were observed in the high-risk patients with IC + CCRT compared to those with IC + IMRT, while comparable outcomes between IC + IMRT and IC + CCRT were shown for low-risk patients. CONCLUSION The radiomics-based nomogram can predict prognosis and survival benefits from concurrent chemotherapy for LANPC following IC. Low-risk patients determined by the nomogram may be potential candidates for omitting concurrent chemotherapy during IMRT. CLINICAL RELEVANCE STATEMENT The radiomics-based nomogram was constructed for risk stratification and patient selection. It can help guide clinical decision-making for patients with locoregionally advanced nasopharyngeal carcinoma following induction chemotherapy, and avoid unnecessary toxicity caused by overtreatment. KEY POINTS • The benefits from concurrent chemotherapy remained controversial for locoregionally advanced nasopharyngeal carcinoma following induction chemotherapy. • Radiomics-based nomogram achieved prognosis and benefits prediction of concurrent chemotherapy. • Low-risk patients defined by the nomogram were candidates for de-intensification.
Collapse
Affiliation(s)
- Shun-Xin Wang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yi Yang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Hui Xie
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Xin Yang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Zhi-Qiao Liu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Hao-Jiang Li
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Wen-Jie Huang
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Wei-Jie Luo
- Department of Medical Oncology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Yi-Ming Lei
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China
| | - Yan-Feng Chen
- Department of Head and Neck Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China.
| | - Li-Zhi Liu
- Department of Radiology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China.
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, 510060, China.
| |
Collapse
|
24
|
Shah A, Dabhade A, Bharadia H, Parekh PS, Yadav MR, Chorawala MR. Navigating the landscape of theranostics in nuclear medicine: current practice and future prospects. Z NATURFORSCH C 2024; 79:235-266. [PMID: 38807355 DOI: 10.1515/znc-2024-0043] [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: 02/25/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024]
Abstract
Theranostics refers to the combination of diagnostic biomarkers with therapeutic agents that share a specific target expressed by diseased cells and tissues. Nuclear medicine is an exciting component explored for its applicability in theranostic concepts in clinical and research investigations. Nuclear theranostics is based on the employment of radioactive compounds delivering ionizing radiation to diagnose and manage certain diseases employing binding with specifically expressed targets. In the realm of personalized medicine, nuclear theranostics stands as a beacon of potential, potentially revolutionizing disease management. Studies exploring the theranostic profile of radioactive compounds have been presented in this review along with a detailed explanation of radioactive compounds and their theranostic applicability in several diseases. It furnishes insights into their applicability across diverse diseases, elucidating the intricate interplay between these compounds and disease pathologies. Light is shed on the important milestones of nuclear theranostics beginning with radioiodine therapy in thyroid carcinomas, MIBG labelled with iodine in neuroblastoma, and several others. Our perspectives have been put forth regarding the most important theranostic agents along with emerging trends and prospects.
Collapse
Affiliation(s)
- Aayushi Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Akshada Dabhade
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Hetvi Bharadia
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Mayur R Yadav
- Department of Pharmacy Practice and Administration, Western University of Health Science, 309 E Second St, Pomona, CA, 91766, USA
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
25
|
Curcean S, Curcean A, Martin D, Fekete Z, Irimie A, Muntean AS, Caraiani C. The Role of Predictive and Prognostic MRI-Based Biomarkers in the Era of Total Neoadjuvant Treatment in Rectal Cancer. Cancers (Basel) 2024; 16:3111. [PMID: 39272969 PMCID: PMC11394290 DOI: 10.3390/cancers16173111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/02/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
The role of magnetic resonance imaging (MRI) in rectal cancer management has significantly increased over the last decade, in line with more personalized treatment approaches. Total neoadjuvant treatment (TNT) plays a pivotal role in the shift from traditional surgical approach to non-surgical approaches such as 'watch-and-wait'. MRI plays a central role in this evolving landscape, providing essential morphological and functional data that support clinical decision-making. Key MRI-based biomarkers, including circumferential resection margin (CRM), extramural venous invasion (EMVI), tumour deposits, diffusion-weighted imaging (DWI), and MRI tumour regression grade (mrTRG), have proven valuable for staging, response assessment, and patient prognosis. Functional imaging techniques, such as dynamic contrast-enhanced MRI (DCE-MRI), alongside emerging biomarkers derived from radiomics and artificial intelligence (AI) have the potential to transform rectal cancer management offering data that enhance T and N staging, histopathological characterization, prediction of treatment response, recurrence detection, and identification of genomic features. This review outlines validated morphological and functional MRI-derived biomarkers with both prognostic and predictive significance, while also exploring the potential of radiomics and artificial intelligence in rectal cancer management. Furthermore, we discuss the role of rectal MRI in the 'watch-and-wait' approach, highlighting important practical aspects in selecting patients for non-surgical management.
Collapse
Affiliation(s)
- Sebastian Curcean
- Department of Radiation Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
- Department of Radiation Oncology, 'Prof. Dr. Ion Chiricuta' Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Andra Curcean
- Department of Imaging, Affidea Center, 15c Ciresilor Street, 400487 Cluj-Napoca, Romania
| | - Daniela Martin
- Department of Radiation Oncology, 'Prof. Dr. Ion Chiricuta' Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Zsolt Fekete
- Department of Radiation Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
- Department of Radiation Oncology, 'Prof. Dr. Ion Chiricuta' Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Alexandru Irimie
- Department of Oncological Surgery and Gynecological Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
- Department of Oncological Surgery, 'Prof. Dr. Ion Chiricuta' Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Alina-Simona Muntean
- Department of Radiation Oncology, 'Prof. Dr. Ion Chiricuta' Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Cosmin Caraiani
- Department of Medical Imaging and Nuclear Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
26
|
Zhang X, Liang X, Wen Y, Wu F, Gao G, Zhang L, Gu Y, Zhang J, Zhou F, Li W, Tang L, Yang X, Zhao H, Zhou C, Hirsch FR. RAC1 inhibition ameliorates IBSP-induced bone metastasis in lung adenocarcinoma. Cell Rep 2024; 43:114528. [PMID: 39052477 DOI: 10.1016/j.celrep.2024.114528] [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: 12/29/2023] [Revised: 05/17/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024] Open
Abstract
Macrophage-to-osteoclast differentiation (osteoclastogenesis) plays an essential role in tumor osteolytic bone metastasis (BM), while its specific mechanisms remain largely uncertain in lung adenocarcinoma BM. In this study, we demonstrate that integrin-binding sialoprotein (IBSP), which is highly expressed in the cancer cells from bone metastatic and primary lesions of patients with lung adenocarcinoma, can facilitate BM and directly promote macrophage-to-osteoclast differentiation independent of RANKL/M-CSF. In vivo results further suggest that osteolytic BM in lung cancer specifically relies on IBSP-induced macrophage-to-osteoclast differentiation. Mechanistically, IBSP regulates the Rac family small GTPase 1 (Rac1)-NFAT signaling pathway and mediates the forward shift of macrophage-to-osteoclast differentiation, thereby leading to early osteolysis. Moreover, inhibition of Rac1 by EHT-1864 or azathioprine in mice models can remarkably alleviate IBSP-induced BM of lung cancer. Overall, our study suggests that tumor-secreted IBSP promotes BM by inducing macrophage-to-osteoclast differentiation, with potential as an early diagnostic maker for BM, and Rac1 can be the therapeutic target for IBSP-promoted BM in lung cancer.
Collapse
Affiliation(s)
- Xiaoshen Zhang
- School of Medicine, Tongji University, Shanghai 200433, China; Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xijun Liang
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai 200433, China
| | - Yaokai Wen
- School of Medicine, Tongji University, Shanghai 200433, China; Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Fengying Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Guanghui Gao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yifeng Gu
- Interventional Radiology Department, Shanghai Sixth People's Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Jianping Zhang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 2000325, China
| | - Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Liang Tang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xiaojun Yang
- Central Laboratory, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Hui Zhao
- Shanghai Sixth People's Hospital affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China.
| | - Fred R Hirsch
- Center of Excellence for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, Box 1128, New York, NY 10029-6574, USA
| |
Collapse
|
27
|
Heidt CM, Bohn JR, Stollmayer R, von Stackelberg O, Rheinheimer S, Bozorgmehr F, Senghas K, Schlamp K, Weinheimer O, Giesel FL, Kauczor HU, Heußel CP, Heußel G. Delta-radiomics features of ADC maps as early predictors of treatment response in lung cancer. Insights Imaging 2024; 15:218. [PMID: 39186132 PMCID: PMC11347553 DOI: 10.1186/s13244-024-01787-5] [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: 02/20/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
OBJECTIVE Investigate the feasibility of detecting early treatment-induced tumor tissue changes in patients with advanced lung adenocarcinoma using diffusion-weighted MRI-derived radiomics features. METHODS This prospective observational study included 144 patients receiving either tyrosine kinase inhibitors (TKI, n = 64) or platinum-based chemotherapy (PBC, n = 80) for the treatment of pulmonary adenocarcinoma. Patients underwent diffusion-weighted MRI the day prior to therapy (baseline, all patients), as well as either + 1 (PBC) or + 7 and + 14 (TKI) days after treatment initiation. One hundred ninety-seven radiomics features were extracted from manually delineated tumor volumes. Feature changes over time were analyzed for correlation with treatment response (TR) according to CT-derived RECIST after 2 months and progression-free survival (PFS). RESULTS Out of 14 selected delta-radiomics features, 6 showed significant correlations with PFS or TR. Most significant correlations were found after 14 days. Features quantifying ROI heterogeneity, such as short-run emphasis (p = 0.04(pfs)/0.005(tr)), gradient short-run emphasis (p = 0.06(pfs)/0.01(tr)), and zone percentage (p = 0.02(pfs)/0.01(tr)) increased in patients with overall better TR whereas patients with worse overall response showed an increase in features quantifying ROI homogeneity, such as normalized inverse difference (p = 0.01(pfs)/0.04(tr)). Clustering of these features allows stratification of patients into groups of longer and shorter survival. CONCLUSION Two weeks after initiation of treatment, diffusion MRI of lung adenocarcinoma reveals quantifiable tissue-level insights that correlate well with future treatment (non-)response. Diffusion MRI-derived radiomics thus shows promise as an early, radiation-free decision-support to predict efficacy and potentially alter the treatment course early. CRITICAL RELEVANCE STATEMENT Delta-Radiomics texture features derived from diffusion-weighted MRI of lung adenocarcinoma, acquired as early as 2 weeks after initiation of treatment, are significantly correlated with RECIST TR and PFS as obtained through later morphological imaging. KEY POINTS Morphological imaging takes time to detect TR in lung cancer, diffusion-weighted MRI might identify response earlier. Several radiomics features are significantly correlated with TR and PFS. Radiomics of diffusion-weighted MRI may facilitate patient stratification and management.
Collapse
Affiliation(s)
- Christian M Heidt
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany.
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.
| | - Jonas R Bohn
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Division of Medical Image Computing, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- National Center for Tumor Diseases (NCT Heidelberg), Heidelberg, Germany
| | - Róbert Stollmayer
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Oyunbileg von Stackelberg
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Stephan Rheinheimer
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Farastuk Bozorgmehr
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Karsten Senghas
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Section for Translational Research, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Kai Schlamp
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Oliver Weinheimer
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Frederik L Giesel
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Hans-Ulrich Kauczor
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Claus Peter Heußel
- Diagnostic and Interventional Radiology, Heidelberg University Hospital, Heidelberg, Germany
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Gudula Heußel
- Translational Lung Research Center (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
- Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
- Pneumology and Respiratory Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
28
|
Katifelis H, Gazouli M. RNA biomarkers in cancer therapeutics: The promise of personalized oncology. Adv Clin Chem 2024; 123:179-219. [PMID: 39181622 DOI: 10.1016/bs.acc.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Cancer therapy is a rapidly evolving and constantly expanding field. Current approaches include surgery, conventional chemotherapy and novel biologic agents as in immunotherapy, that together compose a wide armamentarium. The plethora of choices can, however, be clinically challenging in prescribing the most suitable treatment for any given patient. Fortunately, biomarkers can greatly facilitate the most appropriate selection. In recent years, RNA-based biomarkers have proven most promising. These molecules that range from small noncoding RNAs to protein coding gene transcripts can be valuable in cancer management and especially in cancer therapeutics. Compared to their DNA counterparts which are stable throughout treatment, RNA-biomarkers are dynamic. This allows prediction of success prior to treatment start and can identify alterations in expression that could reflect response. Moreover, improved nucleic acid technology allows RNA to be extracted from practically every biofluid/matrix and evaluated with exceedingly high analytic sensitivity. In addition, samples are largely obtained by minimally invasive procedures and as such can be used serially to assess treatment response real-time. This chapter provides the reader insight on currently known RNA biomarkers, the latest research employing Artificial Intelligence in the identification of such molecules and in clinical decisions driving forward the era of personalized oncology.
Collapse
Affiliation(s)
- Hector Katifelis
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
29
|
Byun H, Han D, Chun HJ, Lee SW. Multiparametric quantification of T1 and T2 relaxation time of bone metastasis in comparison with red or fatty bone marrow using magnetic resonance fingerprinting. Skeletal Radiol 2024; 53:1071-1080. [PMID: 38041749 DOI: 10.1007/s00256-023-04521-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 12/03/2023]
Abstract
OBJECTIVES To assess the T1 and T2 values of bone marrow lesions in spine and pelvis derived from magnetic resonance fingerprinting (MRF) and to evaluate the differences in values among bone metastasis, red marrow and fatty marrow. METHODS Sixty patients who underwent lumbar spine and pelvic MRI with magnetic resonance fingerprinting were retrospectively included. Among eligible patients, those with bone metastasis, benign red marrow deposition and normal fatty marrow were identified. Two radiologists independently measured the T1 and T2 values from metastatic bone lesions, fatty marrow, and red marrow deposition on three-dimensional-magnetic resonance fingerprinting. Intergroup comparison and interobserver agreement were analyzed. RESULTS T1 relaxation time was significantly higher in osteoblastic metastasis than in red marrow (1674.6 ± 436.3 vs 858.7 ± 319.5, p < .001). Intraclass correlation coefficients for T1 and T2 values were 0.96 (p < 0.001) and 0.83 (p < 0.001), respectively. T2 relaxation time of osteoblastic metastasis and red marrow deposition had no evidence of a difference (osteoblastic metastasis, 57.9 ± 25.0 vs red marrow, 58.0 ± 34.4, p = 0.45), as were the average T2 values of osteolytic metastasis and red marrow deposition (osteolytic metastasis, 45.3 ± 15.1 vs red marrow, 58.0 ± 34.4, p = 0.63). CONCLUSIONS We report the feasibility of three-dimensional-magnetic resonance fingerprinting based quantification of bone marrow to differentiate bone metastasis from red marrow. Simultaneous T1 and T2 quantification of metastasis and red marrow deposition was possible in spine and pelvis and showed significant different values with excellent inter-reader agreement. ADVANCE IN KNOWLEDGE T1 values from three-dimensional-magnetic resonance fingerprinting might be a useful quantifier for evaluating bone marrow lesions.
Collapse
Affiliation(s)
- Hokyun Byun
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 1021 Tongil Ro, Eunpyeong-Gu, Seoul, Republic of Korea
| | - Dongyeob Han
- Siemens Healthineers Ltd, Seoul, Republic of Korea
| | - Ho Jong Chun
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-Daero, Seocho-Gu, Seoul, Republic of Korea.
| | - Sheen-Woo Lee
- Department of Radiology, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 1021 Tongil Ro, Eunpyeong-Gu, Seoul, Republic of Korea.
| |
Collapse
|
30
|
Lu XY, Jiang J, Chen S, Qiu YJ, Wang Y, Cheng J, Xu XL, Dong Y, Wang WP. Application of dynamic contrast enhanced ultrasound analysis in predicting early response to systemic therapy of intrahepatic cholangiocarcinoma. Eur J Radiol 2024; 175:111439. [PMID: 38547743 DOI: 10.1016/j.ejrad.2024.111439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/11/2024] [Accepted: 03/20/2024] [Indexed: 05/19/2025]
Abstract
OBJECTIVE To evaluate the value of dynamic contrast-enhanced ultrasound (DCE-US) analysis in early prediction of tumor response to systemic treatment in patients with intrahepatic cholangiocarcinoma (ICC). PATIENTS & METHODS In this retrospective study, patients diagnosed with ICC by core needle biopsy and histopathological results were included. All patients were diagnosed as advanced stages (stage III/IV) by the 8th edition of the American Joint Committee on Cancer (AJCC)/International Union Against Cancer (UICC) TNM staging system. Liver contrast-enhanced ultrasound (CEUS) examination, DCE-US analysis, CT/MRI, and blood tests were performed in all patients before and 2 months after systemic treatment. CEUS procedure was performed using an ultrasound system (ACUSON Sequoia; Siemens Medical Solutions, Germany) equipped with a 5C1 MHz convex array transducer. Time-intensity curves (TIC) and quantitative parameters were created with VueBox software. According to one-year results of the modified Response Evaluation Criteria in Solid Tumors (m-RECIST) based on CT/MRI, patients were divided into the responder group (RG) and the non-responder group (NRG). Before and 2 months after systemic therapy, the DCE-US perfusion parameters was compared using the paired-sample t test and the Wilcoxon test. RESULTS From September 2020 to December 2021, a total of 24 patients diagnosed with advanced ICC were included (11 males, 13 females, mean age 59.4 ± 1.8 years). According to the one year of m-RECIST results, 17 cases (70.8 %) were classified as non-responders by the final m-RECIST criteria, while 7 cases (19.2 %) were responders. Comparing before and 2 months after therapy, the RG took longer time to reach peak intensity, and the peak intensity of TIC was lower. While the TICs of NRG revealed faster enhancement after therapy. Among all DCE-US quantitative parameters, PE (peak enhancement), WiR (wash-in rate), WiPI (wash-in perfusion index) and WoR (wash-out rate) reduced significantly following 2 months of systemic therapy in RG (P < 0.05). Comparing to RG, PE and WiPI decreased slightly 2 months after therapy in NRG (P < 0.05). CONCLUSIONS The DCE-US analysis with quantitative parameters has the potential value to make early and quantitative evaluation of treatment response to systemic therapy in ICC patients.
Collapse
Affiliation(s)
- Xiu-Yun Lu
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Jun Jiang
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Sheng Chen
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Yi-Jie Qiu
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Ying Wang
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Juan Cheng
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Xin-Liang Xu
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Yi Dong
- Department of Ultrasound, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, 200092, Shanghai, China
| | - Wen-Ping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, 200032, Shanghai, China
| |
Collapse
|
31
|
Magbanua MJM, Li W, van ’t Veer LJ. Integrating Imaging and Circulating Tumor DNA Features for Predicting Patient Outcomes. Cancers (Basel) 2024; 16:1879. [PMID: 38791958 PMCID: PMC11120531 DOI: 10.3390/cancers16101879] [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: 04/15/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Biomarkers for evaluating tumor response to therapy and estimating the risk of disease relapse represent tremendous areas of clinical need. To evaluate treatment efficacy, tumor response is routinely assessed using different imaging modalities like positron emission tomography/computed tomography or magnetic resonance imaging. More recently, the development of circulating tumor DNA detection assays has provided a minimally invasive approach to evaluate tumor response and prognosis through a blood test (liquid biopsy). Integrating imaging- and circulating tumor DNA-based biomarkers may lead to improvements in the prediction of patient outcomes. For this mini-review, we searched the scientific literature to find original articles that combined quantitative imaging and circulating tumor DNA biomarkers to build prediction models. Seven studies reported building prognostic models to predict distant recurrence-free, progression-free, or overall survival. Three discussed building models to predict treatment response using tumor volume, pathologic complete response, or objective response as endpoints. The limited number of articles and the modest cohort sizes reported in these studies attest to the infancy of this field of study. Nonetheless, these studies demonstrate the feasibility of developing multivariable response-predictive and prognostic models using regression and machine learning approaches. Larger studies are warranted to facilitate the building of highly accurate response-predictive and prognostic models that are generalizable to other datasets and clinical settings.
Collapse
Affiliation(s)
- Mark Jesus M. Magbanua
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94115, USA;
| | - Wen Li
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA 94115, USA;
| | - Laura J. van ’t Veer
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA 94115, USA;
| |
Collapse
|
32
|
Ueki Y, Otsuka H, Otani T, Kasai R, Otomi Y, Ikemitsu D, Azane S, Kunikane Y, Bando T, Matsuda N, Okada Y, Takayama T, Harada M. Combined visual and quantitative assessment of somatostatin receptor scintigraphy for staging and restaging of neuroendocrine tumors. Jpn J Radiol 2024; 42:519-535. [PMID: 38345724 DOI: 10.1007/s11604-024-01529-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/03/2024] [Indexed: 04/30/2024]
Abstract
PURPOSE Somatostatin receptor scintigraphy (SRS) using 111In-DTPA-DPhe1-octreotide (pentetreotide) has become an integral part of neuroendocrine neoplasm management. The lack of precise quantification is a disadvantage of SRS. This study aimed to adapt the standardized uptake value (SUV) to SRS, establish the SUV range for physiological uptake in the liver, kidney, and spleen, and elucidate the utility of combined visual and quantitative SRS assessment for staging and restaging of neuroendocrine tumors (NETs). MATERIALS AND METHODS This study included 21 patients with NETs who underwent 111In-pentetreotide SRS. The SUV of physiological and pathological uptake was calculated using bone single-photon emission computed tomography (SPECT) quantitative analysis software (GI-BONE). For visual analysis, the primary and metastatic lesions were scored visually on planar and SPECT images using a five-point scale. We assessed the relationships between the SUVs of the liver, kidney, and spleen in the dual phase, and among quantitative indices, visual score, and pathological lesions classification. RESULTS Sixty-three NEN lesions were evaluated. The mean ± standard deviation maximum SUVs (SUVmax) were liver: 4 h, 2.6 ± 1.0; 24 h, 2.2 ± 1.0; kidney: 4 h, 8.9 ± 1.8; 24 h, 7.0 ± 2.0; and spleen; 4 h, 11.3 ± 4.5; 24 h, 11.5 ± 7.6. Higher SUVmax was significantly associated with higher visual scores on dual-phase SPECT (4 h, p < 0.001; 24 h, p < 0.001) (4 h: scores 3 and 4, p < 0.05; scores 3 and 5: p < 0.01; scores 4 and 5: p < 0.01; 24 h: scores 3 and 4, p = 0.0748; scores 3 and 5: p < 0.01; scores 4 and 5: p < 0.01). CONCLUSION We adapted the SUV to SRS and established the range of SUV for physiological uptake in the liver, kidney, and spleen. Combined visual and quantitative assessment is useful for imaging individual lesions in greater detail, and may serve as a new tumor marker of SRS for staging and restaging of NETs.
Collapse
Affiliation(s)
- Yuya Ueki
- Tokushima University Graduate School of Health Sciences, Tokushima, Japan
| | - Hideki Otsuka
- Department of Medical Imaging/Nuclear Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan.
| | - Tamaki Otani
- Advance Radiation Research, Education and Management Center, Tokushima University, Tokushima, Japan
| | - Ryosuke Kasai
- Department of Medical Imaging/Nuclear Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Yoichi Otomi
- Department of Radiology and Radiation Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Daiki Ikemitsu
- Department of Radiology, Tokushima University Hospital, Tokushima, Japan
| | - Shota Azane
- Department of Radiology, Tokushima University Hospital, Tokushima, Japan
| | - Yamato Kunikane
- Department of Radiology, Tokushima University Hospital, Tokushima, Japan
| | - Takanori Bando
- Department of Radiology, Tokushima University Hospital, Tokushima, Japan
| | - Noritake Matsuda
- Department of Radiology, Tokushima University Hospital, Tokushima, Japan
| | - Yasuyuki Okada
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Masafumi Harada
- Department of Radiology and Radiation Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
33
|
da Silva Rosa SC, Barzegar Behrooz A, Guedes S, Vitorino R, Ghavami S. Prioritization of genes for translation: a computational approach. Expert Rev Proteomics 2024; 21:125-147. [PMID: 38563427 DOI: 10.1080/14789450.2024.2337004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 02/21/2024] [Indexed: 04/04/2024]
Abstract
INTRODUCTION Gene identification for genetic diseases is critical for the development of new diagnostic approaches and personalized treatment options. Prioritization of gene translation is an important consideration in the molecular biology field, allowing researchers to focus on the most promising candidates for further investigation. AREAS COVERED In this paper, we discussed different approaches to prioritize genes for translation, including the use of computational tools and machine learning algorithms, as well as experimental techniques such as knockdown and overexpression studies. We also explored the potential biases and limitations of these approaches and proposed strategies to improve the accuracy and reliability of gene prioritization methods. Although numerous computational methods have been developed for this purpose, there is a need for computational methods that incorporate tissue-specific information to enable more accurate prioritization of candidate genes. Such methods should provide tissue-specific predictions, insights into underlying disease mechanisms, and more accurate prioritization of genes. EXPERT OPINION Using advanced computational tools and machine learning algorithms to prioritize genes, we can identify potential targets for therapeutic intervention of complex diseases. This represents an up-and-coming method for drug development and personalized medicine.
Collapse
Affiliation(s)
- Simone C da Silva Rosa
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sofia Guedes
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rui Vitorino
- LAQV/REQUIMTE, Department of Chemistry, University of Aveiro, Aveiro, Portugal
- Department of Medical Sciences, Institute of Biomedicine-iBiMED, University of Aveiro, Aveiro, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, Max Rady College of Medicine, Rady Faculty of Health Science, University of Manitoba, Winnipeg, Canada
- Faculty of Medicine in Zabrze, Academia of Silesia, Katowice, Poland
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
34
|
Nakajima EC, Simpson A, Bogaerts J, de Vries EGE, Do R, Garalda E, Goldmacher G, Kinahan PE, Lambin P, LeStage B, Li Q, Lin F, Litière S, Perez-Lopez R, Petrick N, Schwartz L, Seymour L, Shankar L, Laurie SA. Tumor Size Is Not Everything: Advancing Radiomics as a Precision Medicine Biomarker in Oncology Drug Development and Clinical Care. A Report of a Multidisciplinary Workshop Coordinated by the RECIST Working Group. JCO Precis Oncol 2024; 8:e2300687. [PMID: 38635935 DOI: 10.1200/po.23.00687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 02/08/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Radiomics, the science of extracting quantifiable data from routine medical images, is a powerful tool that has many potential applications in oncology. The Response Evaluation Criteria in Solid Tumors Working Group (RWG) held a workshop in May 2022, which brought together various stakeholders to discuss the potential role of radiomics in oncology drug development and clinical trials, particularly with respect to response assessment. This article summarizes the results of that workshop, reviewing radiomics for the practicing oncologist and highlighting the work that needs to be done to move forward the incorporation of radiomics into clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - Richard Do
- Memorial Sloan-Kettering Cancer Center, NY, NY
| | - Elena Garalda
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | | | | | - Frank Lin
- University of Sydney, Sydney, Australia
| | | | | | | | | | - Lesley Seymour
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Lalitha Shankar
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Scott A Laurie
- The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
35
|
Pourmir I, Van Halteren HK, Elaidi R, Trapani D, Strasser F, Vreugdenhil G, Clarke M. A conceptual framework for cautious escalation of anticancer treatment: How to optimize overall benefit and obviate the need for de-escalation trials. Cancer Treat Rev 2024; 124:102693. [PMID: 38330752 DOI: 10.1016/j.ctrv.2024.102693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND The developmental workflow of the currently performed phase 1, 2 and 3 cancer trial stages lacks essential information required for the determination of the optimal efficacy threshold of new anticancer regimens. Due to this there is a serious risk of overdosing and/or treating for an unnecessary long time, leading to excess toxicity and a higher financial burden for society. But often post-approval de-escalation trials for dose-optimization and treatment de-intensification are not performed due to failing resources and time. Therefore, the developmental workflow needs to be restructured toward cautious systemic cancer treatment escalation, in order to guarantee optimal efficacy and sustainability. METHODS In this manuscript we discuss opportunities to produce the information needed for cautious escalation, based on models of cancer growth and cancer kill kinetics as well as exploratory biomarkers, for the purpose of designing the optimal phase 3 superiority trial. Subsequently, we compare the sample size needed for a phase 3 superiority trial, followed by a necessary de-escalation trial with the sample size needed for a multi-arm phase 3 trial with intervention arms of differing intensity. All essential items are structured within a Framework for Cautious Escalation (FCE). The discussion uses illustrations from the breast cancer setting, but aims to be applicable for all cancers. RESULTS The FCE is a promising model of clinical development in oncology to prevent overtreatment and associated issues, especially with regard to the number of repetitive treatment cycles. It will hopefully increase the relevance and success rate of clinical trials, to deliver improved patient-centric outcomes.
Collapse
Affiliation(s)
- I Pourmir
- Department of Thoracic Oncology, European Hospital Georges Pompidou, Paris, France; INSERM U970, Paris Research Cardiovascular Center, Paris, France
| | - H K Van Halteren
- Department of Medical Oncology, Adrz Hospital, Goes, the Netherlands.
| | - R Elaidi
- Consultant/advisor in Clinical Trials Methodology and Biostatistic, Paris, France
| | - D Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy; Department of Oncology and Haematology, University of Milan, Milan, Italy
| | - F Strasser
- Center for Integrative Medicine, Cantonal Hospital Gallen, St. Gallen University of Bern, Switzerland
| | - G Vreugdenhil
- Department of Medical Oncology, Maxima Medical Center, Veldhoven, the Netherlands
| | - M Clarke
- Professor and Director of Northern Ireland Methodology Hub, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
36
|
Huang L, Kang D, Zhao C, Liu X. Correlation between surrogate endpoints and overall survival in unresectable hepatocellular carcinoma patients treated with immune checkpoint inhibitors: a systematic review and meta-analysis. Sci Rep 2024; 14:4327. [PMID: 38383730 PMCID: PMC10881995 DOI: 10.1038/s41598-024-54945-6] [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: 10/18/2023] [Accepted: 02/19/2024] [Indexed: 02/23/2024] Open
Abstract
This study aimed to assess the therapeutic effect of immune checkpoint inhibitors (ICIs) in patients with unresectable hepatocellular carcinoma (uHCC) and investigate the correlation between surrogate endpoints and overall survival (OS). A systematic literature search included phase I, II, and III clinical trials comparing ICIs to placebo or other therapies for uHCC treatment. Correlations between OS and surrogate endpoints were evaluated using meta-regression analyses and calculating the surrogate threshold effect (STE). The correlation analysis showed a weak association between OS and progression-free survival (PFS), with an R2 value of 0.352 (95% CI: 0.000-0.967). However, complete response (CR) exhibited a strong correlation with OS (R2 = 0.905, 95% CI: 0.728-1.000). Subgroup analyses revealed high correlations between OS and PFS, CR, stable disease (SD), and DC in phase III trials (R2: 0.827-0.922). For the ICI + IA group, significant correlations were observed between OS and SD, progressive disease (PD), and grade 3-5 immune-related adverse events (irAEs) (R2: 0.713-0.969). Analyses of the correlation between survival benefit and risk of mortality across various time points showed a strong association within the first year (R2: 0.724-0.868) but a weak association beyond one year (R2: 0.406-0.499). In ICI trials for uHCC, PFS has limited utility as a surrogate endpoint for OS, while CR exhibits a strong correlation with OS. Subgroup analyses highlight high correlations between OS and PFS, SD, and DC in phase III trials. Notably, the ICI + IA group shows significant associations between OS and SD, PD, and grade 3-5 irAEs. These findings offer valuable insights for interpreting trial outcomes and selecting appropriate endpoints in future clinical studies involving ICIs for uHCC patients.
Collapse
Affiliation(s)
- Litao Huang
- Chinese Evidence-Based Medicine Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Deying Kang
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Evidence-Based Medicine and Clinical Epidemiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chongyang Zhao
- Department of Evidence-Based Medicine and Clinical Epidemiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xueting Liu
- Discipline Construction Department, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
37
|
Sito H, Sharzehan MAK, Islam MA, Tan SC. Genetic Variants Associated With Response to Platinum-Based Chemotherapy in Non-Small Cell Lung Cancer Patients: A Field Synopsis and Meta-Analysis. Br J Biomed Sci 2024; 81:11835. [PMID: 38450253 PMCID: PMC10914946 DOI: 10.3389/bjbs.2024.11835] [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: 07/27/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024]
Abstract
Background: Publications on the associations of genetic variants with the response to platinum-based chemotherapy (PBC) in NSCLC patients have surged over the years, but the results have been inconsistent. Here, a comprehensive meta-analysis was conducted to combine eligible studies for a more accurate assessment of the pharmacogenetics of PBC in NSCLC patients. Methods: Relevant publications were searched in PubMed, Scopus, and Web of Science databases through 15 May 2021. Inclusion criteria for eligible publications include studies that reported genotype and allele frequencies of NSCLC patients treated with PBC, delineated by their treatment response (sensitive vs. resistant). Publications on cell lines or animal models, duplicate reports, and non-primary research were excluded. Epidemiological credibility of cumulative evidence was assessed using the Newcastle-Ottawa Scale (NOS) and Venice criteria. Begg's and Egger's tests were used to assess publication bias. Cochran's Q-test and I2 test were used to calculate the odds ratio and heterogeneity value to proceed with the random effects or fixed-effects method. Venice criteria were used to assess the strength of evidence, replication methods and protection against bias in the studies. Results: A total of 121 publications comprising 29,478 subjects were included in this study, and meta-analyses were performed on 184 genetic variants. Twelve genetic variants from 10 candidate genes showed significant associations with PBC response in NSCLC patients with strong or moderate cumulative epidemiological evidence (increased risk: ERCC1 rs3212986, ERCC2 rs1799793, ERCC2 rs1052555, and CYP1A1 rs1048943; decreased risk: GSTM1 rs36631, XRCC1 rs1799782 and rs25487, XRCC3 rs861539, XPC rs77907221, ABCC2 rs717620, ABCG2 rs2231142, and CDA rs1048977). Bioinformatics analysis predicted possible damaging or deleterious effects for XRCC1 rs1799782 and possible low or medium functional impact for CYP1A1 rs1048943. Conclusion: Our results provide an up-to-date summary of the association between genetic variants and response to PBC in NSCLC patients.
Collapse
Affiliation(s)
- Hilary Sito
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | | | - Md Asiful Islam
- WHO Collaborating Centre for Global Women’s Health, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Sathish G, Monavarshini LK, Sundaram K, Subramanian S, Kannayiram G. Immunotherapy for lung cancer. Pathol Res Pract 2024; 254:155104. [PMID: 38244436 DOI: 10.1016/j.prp.2024.155104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/22/2024]
Abstract
Immune checkpoint blockers have transformed non-small-cell lung cancer treatment, but they can lead to autoimmune and inflammatory side effects, leading to the concurrent use of immunosuppressive treatments. In this analysis, we delve into the potential of antibodies checkpoint blockade, focusing on CTLA-4 inhibition using ipilimumab, as a groundbreaking cancer immunotherapy. We also concentrate on the role of biomarkers, particularly PD-L1 activity and mutation significance, in predicting the response to programmed cell death protein 1 blockage and the prevalence of side effects associated with immune-related side effects. In describing the patterns of cancer response to immunotherapy, we underline the limitations of response assessment criteria like RECIST and World Health Organization. We also stress the necessity of ongoing studies and clinical trials, standardized guidelines, and additional research to improve response assessment in the era of immunotherapy.
Collapse
Affiliation(s)
- Girshani Sathish
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - L K Monavarshini
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Keerthi Sundaram
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Sendilvelan Subramanian
- Deparment of Mechanical Engineering, Dr.MGR Educational and Research Institute, Maduravoyal, Chennai 600095, India
| | - Gomathi Kannayiram
- Department of Biotechnology, Dr. M.G.R. Educational and Research Institute, Maduravoyal, Chennai 600095, India.
| |
Collapse
|
39
|
Danzer MF, Eveslage M, Görlich D, Noto B. A statistical framework for planning and analysing test-retest studies of repeatability. Stat Methods Med Res 2024; 33:295-308. [PMID: 38298010 DOI: 10.1177/09622802241227959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
There is an increasing number of potential quantitative biomarkers that could allow for early assessment of treatment response or disease progression. However, measurements of such biomarkers are subject to random variability. Hence, differences of a biomarker in longitudinal measurements do not necessarily represent real change but might be caused by this random measurement variability. Before utilizing a quantitative biomarker in longitudinal studies, it is therefore essential to assess the measurement repeatability. Measurement repeatability obtained from test-retest studies can be quantified by the repeatability coefficient, which is then used in the subsequent longitudinal study to determine if a measured difference represents real change or is within the range of expected random measurement variability. The quality of the point estimate of the repeatability coefficient, therefore, directly governs the assessment quality of the longitudinal study. Repeatability coefficient estimation accuracy depends on the case number in the test-retest study, but despite its pivotal role, no comprehensive framework for sample size calculation of test-retest studies exists. To address this issue, we have established such a framework, which allows for flexible sample size calculation of test-retest studies, based upon newly introduced criteria concerning assessment quality in the longitudinal study. This also permits retrospective assessment of prior test-retest studies.
Collapse
Affiliation(s)
- Moritz Fabian Danzer
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Maria Eveslage
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Dennis Görlich
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Benjamin Noto
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
- Clinic for Radiology, University Hospital Münster, Münster, Germany
- Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
40
|
Zhu HB, Zhao B, Li XT, Zhang XY, Yao Q, Sun YS. Value of multiple models of diffusion-weighted imaging to predict hepatic lymph node metastases in colorectal liver metastases patients. World J Gastroenterol 2024; 30:308-317. [PMID: 38313236 PMCID: PMC10835543 DOI: 10.3748/wjg.v30.i4.308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND About 10%-31% of colorectal liver metastases (CRLM) patients would concomitantly show hepatic lymph node metastases (LNM), which was considered as sign of poor biological behavior and a relative contraindication for liver resection. Up to now, there's still lack of reliable preoperative methods to assess the status of hepatic lymph nodes in patients with CRLM, except for pathology examination of lymph node after resection. AIM To compare the ability of mono-exponential, bi-exponential, and stretched-exponential diffusion-weighted imaging (DWI) models in distinguishing between benign and malignant hepatic lymph nodes in patients with CRLM who received neoadjuvant chemotherapy prior to surgery. METHODS In this retrospective study, 97 CRLM patients with pathologically confirmed hepatic lymph node status underwent magnetic resonance imaging, including DWI with ten b values before and after chemotherapy. Various parameters, such as the apparent diffusion coefficient from the mono-exponential model, and the true diffusion coefficient, the pseudo-diffusion coefficient, and the perfusion fraction derived from the intravoxel incoherent motion model, along with distributed diffusion coefficient (DDC) and α from the stretched-exponential model (SEM), were measured. The parameters before and after chemotherapy were compared between positive and negative hepatic lymph node groups. A nomogram was constructed to predict the hepatic lymph node status. The reliability and agreement of the measurements were assessed using the coefficient of variation and intraclass correlation coefficient. RESULTS Multivariate analysis revealed that the pre-treatment DDC value and the short diameter of the largest lymph node after treatment were independent predictors of metastatic hepatic lymph nodes. A nomogram combining these two factors demonstrated excellent performance in distinguishing between benign and malignant lymph nodes in CRLM patients, with an area under the curve of 0.873. Furthermore, parameters from SEM showed substantial repeatability. CONCLUSION The developed nomogram, incorporating the pre-treatment DDC and the short axis of the largest lymph node, can be used to predict the presence of hepatic LNM in CRLM patients undergoing chemotherapy before surgery. This nomogram was proven to be more valuable, exhibiting superior diagnostic performance compared to quantitative parameters derived from multiple b values of DWI. The nomogram can serve as a preoperative assessment tool for determining the status of hepatic lymph nodes and aiding in the decision-making process for surgical treatment in CRLM patients.
Collapse
Affiliation(s)
- Hai-Bin Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Bo Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiao-Ting Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiao-Yan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Qian Yao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Ying-Shi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
41
|
Lee H, Ahn TR, Hwang KH, Lee SW. Evaluation of Three Imaging Methods to Quantify Key Events in Pelvic Bone Metastasis. Cancers (Basel) 2024; 16:214. [PMID: 38201641 PMCID: PMC10778360 DOI: 10.3390/cancers16010214] [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: 11/14/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The purpose of this study is to compare turbo spin echo diffusion-weighted images in radial trajectory (BLADE DWI) with multi-shot echoplanar imaging (RESOLVE DWI) for imaging the metastatic lesion in the pelvic bone to find a correlation between ADC values and standardized uptake values (SUVs) of FDG uptake in PET/CT. The study also seeks to compare the values of metastatic lesions with those of benign bone lesions, specifically red marrow hyperplasia. METHODS The retrospective IRB-approved study included patients with bone metastasis and red marrow hyperplasia in the pelvic bone who underwent 3.0 T MRI with BLADE/RESOLVE DWI sequences and F-18 FDG PET/CT within one month. BVC (best value comparator) was used in determining the nature of bone lesions. Apparent diffusion coefficient (ADC) and standardized uptake value (SUV) were measured by a radiologist and a nuclear medicine physician. MRI image quality was graded with a Likert scale regarding the visualization of the sacroiliac joint, sacral neural foramen, hamstring tendon at ischial tuberosity, and tumor border. Signal-to-noise ratio (SNR) and imaging time were compared between the two DWIs. Mean, peak, and maximum SUVs between metastatic and benign red marrow lesions were compared. SUVs and ADC values were compared. AUROC analyses and cut-off values were obtained for each parameter. Mann-Whitney U, Spearman's rho, and Kolmogorov-Smirnov tests were applied using SPSS. RESULTS The final study group included 58 bone lesions (19 patients (male: female = 6:13, age 52.5 ± 9.6, forty-four (75.9%) bone metastasis, fourteen (24.1%) benign red marrow hyperplasia). ADCs from BLADE and RESOLVE were significantly higher in bone metastasis than red marrow hyperplasia. BLADE showed higher ADC values, higher anatomical scores, and higher SNR than RESOLVE DWI (p < 0.05). Imaging times were longer for BLADE than RESOLVE (6 min 3 s vs. 3 min 47 s, p < 0.05). There was a poor correlation between ADC values and SUVs (correlation coefficient from 0.04 to 0.31). The AUROC values of BLADE and RESOLVE MRI ranged from 0.892~0.995. Those of PET ranged from 0.877~0.895. The cut-off ADC values between the bone metastasis and red marrow hyperplasia were 355.0, 686.5, 531.0 for BLADE min, max, and average, respectively, and 112.5, 737.0, 273.0 for RESOLVE min, max, and average, respectively. The cut-off SUV values were 1.84, 5.01, and 3.81 for mean, peak, and max values, respectively (p < 0.05). CONCLUSIONS Compared with RESOLVE DWI, BLADE DWI showed improved image quality of pelvic bone MRI in the aspect of anatomical depiction and SNR, higher ADC values, albeit longer imaging time. BLADE and RESOLVE could differentiate bone metastasis and red marrow hyperplasia with quantifiable cut-off values. Further study is necessary to evaluate the discrepancy between the quantifiers between PET and MRI.
Collapse
Affiliation(s)
- Haejun Lee
- Department of Nuclear Medicine, Gachon University Gil Hospital, Incheon 21565, Republic of Korea; (H.L.); (K.H.H.)
| | - Tae Ran Ahn
- Department of Radiology, Gachon University Gil Hospital, Incheon 21565, Republic of Korea;
| | - Kyung Hoon Hwang
- Department of Nuclear Medicine, Gachon University Gil Hospital, Incheon 21565, Republic of Korea; (H.L.); (K.H.H.)
| | - Sheen-Woo Lee
- Department of Radiology, The Catholic University of Korea Eunpyeong St. Mary’s Hospital, Seoul 03312, Republic of Korea
| |
Collapse
|
42
|
Li Y, Zhang H, Yue L, Fu C, Grimm R, Li W, Guo W, Tong T. Whole tumor based texture analysis of magnetic resonance diffusion imaging for colorectal liver metastases: A prospective study for diffusion model comparison and early response biomarker. Eur J Radiol 2024; 170:111203. [PMID: 38007855 DOI: 10.1016/j.ejrad.2023.111203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/16/2023] [Accepted: 11/14/2023] [Indexed: 11/28/2023]
Abstract
PURPOSE To evaluate and compare the diagnostic value of diffusion-related texture analysis parameters obtained from various magnetic resonance diffusion models as early predictors of the clinical response to chemotherapy in patients with colorectal liver metastases (CRLM). METHODS Patients (n = 145) with CRLM were prospectively and consecutively enrolled and scanned using diffusion-weighted imaging (DWI)-magnetic resonance imaging (MRI)/intravoxel incoherent motion (IVIM)/diffusion kurtosis imaging (DKI) before (baseline) and two-three weeks after (follow-up) commencing chemotherapy. Therapy response was evaluated based on the Response Evaluation Criteria in Solid Tumors (RECIST, version 1.1). The histogram and texture parameters of each diffusion-related parametric map were analysed between the responding and non-responding groups, screened using LASSO, and fitted with binary logistic regression models. The diagnostic efficacy of each model in the early prediction of CRLM was analysed, and the corresponding receiver operating characteristic (ROC) curve was drawn. The area under the curve (AUC) and 95% confidence intervals (CI) were calculated. RESULTS Of the 145 analysed patients, 69 were in the responding group and 76 were in the non-responding group. Among all models, the difference value based on the histogram and texture features of the DKI-derived parameters performed best for the early prediction of CRLM treatment efficacy. The AUC of the DKI model in the validation set reached 0.795 (95% CI 0.652-0.938). Among the IVIM-derived parameters, the difference model based on D and D* performed best, and the AUC in the validation set reached 0.737 (95% CI 0.586-0.889). Finally, in the DWI sequence, the model comprising baseline features performed the best, with an AUC of 0.699 (95% CI 0.537-0.86) in the validation set. CONCLUSIONS Baseline DWI parameters and follow-up changes in IVIM and DKI parameters predicted the chemotherapeutic response in patients with CRLM. In addition, as very early predictors, DKI-derived parameters were more effective than DWI- and IVIM-related parameters, in which changes in D-parameters performed best.
Collapse
Affiliation(s)
- Yue Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Yue
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Caixia Fu
- MR Collaboration, Siemens Shenzhen Magnetic Resonance Ltd., Shenzhen, China
| | - Robert Grimm
- MR Application Predevelopment, Siemens Healthcare GmbH, Erlangen, Germany
| | - Wenhua Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Weijian Guo
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Tong Tong
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
43
|
Trulson I, Holdenrieder S. Prognostic value of blood-based protein biomarkers in non-small cell lung cancer: A critical review and 2008-2022 update. Tumour Biol 2024; 46:S111-S161. [PMID: 37927288 DOI: 10.3233/tub-230009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Therapeutic possibilities for non-small cell lung cancer (NSCLC) have considerably increased during recent decades. OBJECTIVE To summarize the prognostic relevance of serum tumor markers (STM) for early and late-stage NSCLC patients treated with classical chemotherapies, novel targeted and immune therapies. METHODS A PubMed database search was conducted for prognostic studies on carcinoembryonic antigen (CEA), cytokeratin-19 fragment (CYFRA 21-1), neuron-specific enolase, squamous-cell carcinoma antigen, progastrin-releasing-peptide, CA125, CA 19-9 and CA 15-3 STMs in NSCLC patients published from 2008 until June 2022. RESULTS Out of 1069 studies, 141 were identified as meeting the inclusion criteria. A considerable heterogeneity regarding design, patient number, analytical and statistical methods was observed. High pretherapeutic CYFRA 21-1 levels and insufficient decreases indicated unfavorable prognosis in many studies on NSCLC patients treated with chemo-, targeted and immunotherapies or their combinations in early and advanced stages. Similar results were seen for CEA in chemotherapy, however, high pretherapeutic levels were sometimes favorable in targeted therapies. CA125 is a promising prognostic marker in patients treated with immunotherapies. Combinations of STMs further increased the prognostic value over single markers. CONCLUSION Protein STMs, especially CYFRA 21-1, have prognostic potential in early and advanced stage NSCLC. For future STM investigations, better adherence to comparable study designs, analytical methods, outcome measures and statistical evaluation standards is recommended.
Collapse
Affiliation(s)
- Inga Trulson
- Munich Biomarker Research Center, Institute for Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
| | - Stefan Holdenrieder
- Munich Biomarker Research Center, Institute for Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
| |
Collapse
|
44
|
Luo W, Xiu Z, Wang X, McGarry R, Allen J. A Novel Method for Evaluating Early Tumor Response Based on Daily CBCT Images for Lung SBRT. Cancers (Basel) 2023; 16:20. [PMID: 38201447 PMCID: PMC10778260 DOI: 10.3390/cancers16010020] [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: 11/03/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND We aimed to develop a new tumor response assessment method for lung SBRT. METHODS In total, 132 lung cancer patients with 134 tumors who received SBRT treatment with daily CBCT were included in this study. The information about tumor size (area), contrast (contrast-to-noise ratio (CNR)), and density/attenuation (μ) was derived from the CBCT images for the first and the last fractions. The ratios of tumor area, CNR, and μ (RA, RCNR, Rμ) between the last and first fractions were calculated for comparison. The product of the three rations was defined as a new parameter (R) for assessment. Tumor response was independently assessed by a radiologist based on a comprehensive analysis of the CBCT images. RESULTS R ranged from 0.27 to 1.67 with a mean value of 0.95. Based on the radiologic assessment results, a receiver operation characteristic (ROC) curve with the area under the curve (AUC) of 95% was obtained and the optimal cutoff value (RC) was determined as 1.1. The results based on RC achieved a 94% accuracy, 94% specificity, and 90% sensitivity. CONCLUSION The results show that R was correlated with early tumor response to lung SBRT and that using R for evaluating tumor response to SBRT would be viable and efficient.
Collapse
Affiliation(s)
- Wei Luo
- Department of Radiation Medicine, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA; (Z.X.); (R.M.)
| | - Zijian Xiu
- Department of Radiation Medicine, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA; (Z.X.); (R.M.)
| | - Xiaoqin Wang
- Department of Radiology, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA;
| | - Ronald McGarry
- Department of Radiation Medicine, University of Kentucky, 800 Rose Street, Lexington, KY 40536, USA; (Z.X.); (R.M.)
| | - Joshua Allen
- AdventHealth, 2501 N Orange Ave, Orlando, FL 32804, USA;
| |
Collapse
|
45
|
Tao W, Wang W, Zhai J, Guo L. Efficacy Analysis of Neoadjuvant versus Adjuvant Cisplatin-Paclitaxel Regimens for Initial Treatment of FIGO Stages IB3 and IIA2 Cervical Cancer. Med Sci Monit 2023; 29:e940545. [PMID: 38062672 PMCID: PMC10714867 DOI: 10.12659/msm.940545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Large cancer lesions are often challenging to treat with surgical intervention alone. Neoadjuvant chemotherapy is frequently used for FIGO stage IB3 and IIA2 cervical cancers to optimize the outcomes of radical surgeries. This study aimed to compare the effectiveness of neoadjuvant chemotherapy, followed by adjuvant chemotherapy and radiotherapy, if necessary, with the traditional approach of adjuvant chemotherapy and radiotherapy after radical hysterectomy in treatment-naïve patients with cervical cancer of specified stages. MATERIAL AND METHODS A total of 245 female patients were administered either 70 to 85 mg/m² cisplatin and 165 to 175 mg/m² paclitaxel every 21 days (2 cycles) prior to radical hysterectomy, followed by adjuvant chemotherapy and radiotherapy if needed (neoadjuvant therapy, NT cohort, n=105), or received adjuvant chemotherapy and radiotherapy after radical hysterectomy adjuvant therapy, AT cohort, n=140). RESULTS In the NT cohort, 76% of patients responded to neoadjuvant chemotherapy, while 24% did not. Adverse operative, intraoperative, and postoperative outcomes were significantly more common among the non-responders (P<0.05). After 5 years, 91% of responders and 72% of non-responders survived without recurrence (P=0.0372), and 3% of responders and 28% of non-responders had died (P=0.0005). CONCLUSIONS The resistance to neoadjuvant chemotherapy is a poor prognostic factor. Neoadjuvant chemotherapy followed by radical hysterectomy and adjuvant chemotherapy/radiotherapy appears to be advantageous for cervical cancer patients who respond well to neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Wei Tao
- Department of Gynecology, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| | - Weiqi Wang
- Department of Gynecology, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| | - Jingfang Zhai
- Prenatal Diagnosis Medical Center, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| | - Linlin Guo
- Department of Gynecology, Xuzhou Central Hospital, Xuzhou, Jiangsu, PR China
| |
Collapse
|
46
|
Liu M, Li X, Zhang H, Ren F, Liu J, Li Y, Dong M, Zhao H, Xu S, Liu H, Chen J. Apatinib added when NSCLC patients get slow progression with EGFR-TKI: A prospective, single-arm study. Cancer Med 2023; 12:21735-21741. [PMID: 38033095 PMCID: PMC10757148 DOI: 10.1002/cam4.6737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/17/2023] [Accepted: 11/07/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKI) acquired resistance was an inevitably events in NSCLC treatment. AIMS Intending to overcome the acquired resistance of EGFR-TKI. MATERIALS & METHODS A clinical trial was, we enrolled 12 patients who were slowly progressing on first-generation EGFR-TKI, and added apatinib when the patients got slow progression. RESULTS Seven patients were included in the efficacy analysis. The median PFS2 of apatinib combined with EGFR-TKI was 8.2 months (95% CI, 7.3 m-NA), and the total PFS reached 20.9 months (95% CI, 17.3 m-NA) when plus PFS1. All the adverse events were manageable. The median PFS was significantly longer for circulating tumor DNA (ctDNA)-cleared patients (8.4 months; 95% CI, 8.2-NA) than for those ctDNA not cleared (7.1 months; 95% CI, 6.9-NA) (p = 0.0082). DISCUSSION The addition of apatinib did improve the duration of first-generation EGFR-TKI use, and the duration was better than the first-line use of third-generation EGFR-TKI. CONCLUSION The addition of apatinib when the patients got slow progression after initial EGFR-TKI therapy may be a good treatment option and the side effects are controllable. It is possible to monitor treatment efficacy using ctDNA.
Collapse
Affiliation(s)
- Minghui Liu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Xin Li
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Hongbing Zhang
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Fan Ren
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Jinghao Liu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Yongwen Li
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentTianjin Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Ming Dong
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Honglin Zhao
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Song Xu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentTianjin Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| | - Jun Chen
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor MicroenvironmentTianjin Lung Cancer Institute, Tianjin Medical University General HospitalTianjinChina
| |
Collapse
|
47
|
Swinburne NC, Yadav V, Murthy KNK, Elnajjar P, Shih HH, Panyam PK, Santilli A, Gutman DC, Pike L, Moss NS, Stone J, Hatzoglou V, Shah A, Juluru K, Shah SP, Holodny AI, Young RJ. Fast, light, and scalable: harnessing data-mined line annotations for automated tumor segmentation on brain MRI. Eur Radiol 2023; 33:6582-6591. [PMID: 37042979 PMCID: PMC10523913 DOI: 10.1007/s00330-023-09583-3] [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: 10/04/2022] [Revised: 02/04/2023] [Accepted: 02/16/2023] [Indexed: 04/13/2023]
Abstract
OBJECTIVES While fully supervised learning can yield high-performing segmentation models, the effort required to manually segment large training sets limits practical utility. We investigate whether data mined line annotations can facilitate brain MRI tumor segmentation model development without requiring manually segmented training data. METHODS In this retrospective study, a tumor detection model trained using clinical line annotations mined from PACS was leveraged with unsupervised segmentation to generate pseudo-masks of enhancing tumors on T1-weighted post-contrast images (9911 image slices; 3449 adult patients). Baseline segmentation models were trained and employed within a semi-supervised learning (SSL) framework to refine the pseudo-masks. Following each self-refinement cycle, a new model was trained and tested on a held-out set of 319 manually segmented image slices (93 adult patients), with the SSL cycles continuing until Dice score coefficient (DSC) peaked. DSCs were compared using bootstrap resampling. Utilizing the best-performing models, two inference methods were compared: (1) conventional full-image segmentation, and (2) a hybrid method augmenting full-image segmentation with detection plus image patch segmentation. RESULTS Baseline segmentation models achieved DSC of 0.768 (U-Net), 0.831 (Mask R-CNN), and 0.838 (HRNet), improving with self-refinement to 0.798, 0.871, and 0.873 (each p < 0.001), respectively. Hybrid inference outperformed full image segmentation alone: DSC 0.884 (Mask R-CNN) vs. 0.873 (HRNet), p < 0.001. CONCLUSIONS Line annotations mined from PACS can be harnessed within an automated pipeline to produce accurate brain MRI tumor segmentation models without manually segmented training data, providing a mechanism to rapidly establish tumor segmentation capabilities across radiology modalities. KEY POINTS • A brain MRI tumor detection model trained using clinical line measurement annotations mined from PACS was leveraged to automatically generate tumor segmentation pseudo-masks. • An iterative self-refinement process automatically improved pseudo-mask quality, with the best-performing segmentation pipeline achieving a Dice score of 0.884 on a held-out test set. • Tumor line measurement annotations generated in routine clinical radiology practice can be harnessed to develop high-performing segmentation models without manually segmented training data, providing a mechanism to rapidly establish tumor segmentation capabilities across radiology modalities.
Collapse
Affiliation(s)
- Nathaniel C Swinburne
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA.
| | - Vivek Yadav
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | | | - Pierre Elnajjar
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Hao-Hsin Shih
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Prashanth Kumar Panyam
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Alice Santilli
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - David C Gutman
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Luke Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nelson S Moss
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacqueline Stone
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vaios Hatzoglou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Akash Shah
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Krishna Juluru
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrei I Holodny
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| | - Robert J Young
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, 10065, USA
| |
Collapse
|
48
|
Chien HC, Yeh LR, Hung KC, Lim SW, Cheng CY, Lee YC, Chen JH, Ko CC. Pretreatment diffusion-weighted imaging for prediction of relapsed and refractory primary central nervous system lymphoma. Front Neurol 2023; 14:1227607. [PMID: 37638189 PMCID: PMC10447899 DOI: 10.3389/fneur.2023.1227607] [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: 05/23/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Objectives A subset of primary central nervous system lymphoma (PCNSL) has been shown to undergo an early relapsed/refractory (R/R) period after first-line chemotherapy. This study investigated the pretreatment clinical and MRI features to predict R/R in PCNSL, emphasizing the apparent diffusion coefficient (ADC) values in diffusion-weighted imaging (DWI). Methods This retrospective study investigated the pretreatment MRI features for predicting R/R in PCNSL. Only patients who had undergone complete preoperative and postoperative MRI follow-up studies were included. From January 2006 to December 2021, 52 patients from two medical institutions with a diagnosis of PCNSL were included (median follow-up time, 26.3 months). Among these, 24 (46.2%) had developed R/R (median time to relapse, 13 months). Cox proportional hazard regression analyses were performed to determine hazard ratios for all parameters. Results Significant predictors of R/R in PCNSL were female sex, complete response (CR) to first-line chemotherapy, and ADC value/ratio (p < 0.05). Cut-off points of ADC values and ADC ratios for prediction of R/R were 0.68 × 10-3 mm2/s and 0.97, with AUCs of 0.78 and 0.77, respectively (p < 0.05). Multivariate Cox proportional hazards analysis showed that failure of CR to first-line chemotherapy and low ADC values (<0.68 × 10-3 mm2/s) were significant risk factors for R/R, with hazard ratios of 5.22 and 14.45, respectively (p < 0.05). Kaplan-Meier analysis showed that lower ADC values and ratios predicted significantly shorter progression-free survival (p < 0.05). Conclusion Pretreatment ADC values in DWI offer quantitative valuable information for the treatment planning in PCNSL.
Collapse
Affiliation(s)
- Hsi-Cheng Chien
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan
| | - Lee-Ren Yeh
- Department of Medical Imaging, E-Da Hospital, Kaohsiung, Taiwan
- Department of Medical Imaging and Radiological Sciences, College of Medicine, I-Shou University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Kuo-Chuan Hung
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Hospital and Health Care Administration, College of Recreation and Health Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Sher-Wei Lim
- Department of Neurosurgery, Chi Mei Medical Center, Chiali, Tainan, Taiwan
- Department of Nursing, Min-Hwei College of Health Care Management, Tainan, Taiwan
| | - Chung-Yu Cheng
- Department of Medical Imaging, E-Da Hospital, Kaohsiung, Taiwan
| | - Yu-Chang Lee
- Department of Medical Imaging, E-Da Hospital, Kaohsiung, Taiwan
| | - Jeon-Hor Chen
- Department of Medical Imaging, E-Da Hospital, Kaohsiung, Taiwan
- Department of Radiological Sciences, University of California, Irvine, Irvine, CA, United States
| | - Ching-Chung Ko
- Department of Medical Imaging, Chi Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
49
|
Criss CR, Makary MS. Recent Advances in Image-Guided Locoregional Therapies for Primary Liver Tumors. BIOLOGY 2023; 12:999. [PMID: 37508428 PMCID: PMC10376862 DOI: 10.3390/biology12070999] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
Primary liver cancer is the leading cause of cancer-related deaths worldwide. with incidences predicted to rise over the next several decades. Locoregional therapies, such as radiofrequency or microwave ablation, are described as image-guided percutaneous procedures, which offer either a curative intent for early-stage hepatocellular carcinoma or bridging/downstaging for surgical resection or transplantation. Catheter-driven locoregional therapies, such as transarterial chemoembolization and radioembolization, induce tumor hypoxia, can be palliative, and improve survival for early-to-intermediate hepatocellular carcinoma and unresectable intrahepatic cholangiocarcinoma. Herein, we provide a comprehensive overview of the antineoplastic mechanisms underpinning locoregional therapies, different treatment approaches, and the current state of the literature for the efficacy of locoregional therapies for primary liver cancer. We also discuss emerging advancements, such as the adjuvant use of immunotherapies and molecular targeting agents with locoregional therapy, for the treatment of primary liver cancer.
Collapse
Affiliation(s)
- Cody R. Criss
- OhioHealth Riverside Methodist Hospital, Columbus, OH 43214, USA;
| | - Mina S. Makary
- Department of Radiology, The Ohio State University Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
50
|
Khan F, Jones K, Lyon P. Immune checkpoint inhibition: a future guided by radiology. Br J Radiol 2023; 96:20220565. [PMID: 36752570 PMCID: PMC10321249 DOI: 10.1259/bjr.20220565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 01/04/2023] [Accepted: 01/29/2023] [Indexed: 02/09/2023] Open
Abstract
The limitation of the function of antitumour immune cells is a common hallmark of cancers that enables their survival. As such, the potential of immune checkpoint inhibition (ICI) acts as a paradigm shift in the treatment of a range of cancers but has not yet been fully capitalised. Combining minimally and non-invasive locoregional therapies offered by radiologists with ICI is now an active field of research with the aim of furthering therapeutic capabilities in medical oncology. In parallel to this impending advancement, the "imaging toolbox" available to radiologists is also growing, enabling more refined tumour characterisation as well as greater accuracy in evaluating responses to therapy. Options range from metabolite labelling to cellular localisation to immune checkpoint screening. It is foreseeable that these novel imaging techniques will be integrated into personalised treatment algorithms. This growth in the field must include updating the current standardised imaging criteria to ensure they are fit for purpose. Such criteria is crucial to both appropriately guide clinical decision-making regarding next steps of treatment, but also provide reliable prognosis. Quantitative approaches to these novel imaging techniques are also already being investigated to further optimise personalised therapeutic decision-making. The therapeutic potential of specific ICIs and locoregional therapies could be determined before administration thus limiting unnecessary side-effects whilst maintaining efficacy. Several radiological aspects of oncological care are advancing simultaneously. Therefore, it is essential that each development is assessed for clinical use and optimised to ensure the best treatment decisions are being offered to the patient. In this review, we discuss state of the art advances in novel functional imaging techniques in the field of immuno-oncology both pre-clinically and clinically.
Collapse
Affiliation(s)
- Faraaz Khan
- Foundation Doctor, Buckinghamshire Hospitals NHS Trust, Amersham, Buckinghamshire, United Kingdom
| | - Keaton Jones
- Academic Clinical Lecturer Nuffield Department of Surgical Sciences University of Oxford, Wellington Square, Oxford, United Kingdom
| | - Paul Lyon
- Consultant Radiologist, Department of Radiology, Oxford University Hospitals, Headington, Oxford, United Kingdom
| |
Collapse
|