1
|
Chen J, Ma Z, Xu Y, Ge J, Yao H, Li C, Hu X, Pu Y, Li M, Jiang C. CT-based machine learning radiomics predicts Ki-67 expression level and its relationship with overall survival in resectable pancreatic ductal adenocarcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04798-y. [PMID: 39841230 DOI: 10.1007/s00261-025-04798-y] [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: 11/13/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Abstract
BACKGROUND The prognostic prediction of pancreatic ductal adenocarcinoma (PDAC) remains challenging. This study aimed to develop a radiomics model to predict Ki-67 expression status in PDAC patients using radiomics features from dual-phase enhanced CT, and integrated clinical characteristics to create a radiomics-clinical nomogram for prognostic prediction. METHODS In this retrospective study, data were collected from 124 PDAC patients treated surgically at a single center, from January 2017 to March 2023. Patients were categorized according to the Ki-67 expression rate. Radiomics features were extracted from arterial and portal venous phase CT images using 3D Slicer v5.0.3. A radiomics model was formulated and validated to predict the Ki-67 expression, and a nomogram combining clinical indicators and the radiomics model was developed to predict 1, 2 and 3 year overall survival (OS). RESULTS The optimal Ki-67 expression rate cutoff was identified as 50%, with significant OS differences. The developed radiomics model showed good predictive ability with area under the curves of 0.806 and 0.801 in the training and validation groups, respectively. High radiomics score, elevated carbohydrate antigen 19-9 (CA19-9), and receipt of adjuvant chemotherapy were identified as independent prognostic factors for OS. The radiomics-clinical nomogram accurately predicted 1, 2 and 3 year OS in PDAC patients. CONCLUSIONS The radiomics-clinical nomogram provides a non-invasive and efficient method for predicting Ki-67 expression and overall survival in PDAC patients, which could potentially guide clinical decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Zhuangxuan Ma
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yamin Xu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Jieqiong Ge
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Hongfei Yao
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Chunjing Li
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Xiao Hu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Yunlong Pu
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Chongyi Jiang
- Department of Hepato-Biliary-Pancreatic Surgery, Huadong Hospital affiliated to Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Kang YK, Ha S, Jeong JB, Oh SW. The value of PET/CT radiomics for predicting survival outcomes in patients with pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:28958. [PMID: 39578496 PMCID: PMC11584753 DOI: 10.1038/s41598-024-77022-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis even without distant metastases, necessitating in-depth characterization of primary tumors for survival prediction. We assessed the feasibility of using FDG-PET/CT radiomics to predict overall survival (OS) in PDAC. This retrospective study included PDAC patients without distant metastasis who underwent FDG-PET/CT for initial staging. Primary tumors were segmented from FDG-PET/CT images, extracting 222 radiomics features. A radiomics-based risk score (Rad-score) was developed using Cox proportional hazards regression with LASSO to predict OS. The prognostic performance of the Rad-score was compared with a clinical model (demographics, disease stage, laboratory results) using Harrell's concordance index (C-index) and bootstrapping. 140 patients were included, with a mortality rate was 72.9% during follow-up (total population, 19.5 ± 19.2 months; survivors, 34.4 ± 28.8 months). Eleven radiomics features were significant for survival prediction. The Rad-score predicted OS with a C-index of 0.681 [95% CI, 0.632-0.731]. A model integrating clinical parameters and Rad-score outperformed the clinical-only model in predicting OS (C-index 0.740 [0.715-0.816] vs. 0.673 [0.650-0.766]; C-index difference 0.067 [0.014-0.113]; P < 0.001). These findings suggest that incorporating FDG-PET/CT radiomics into preexisting prognotic stratification paradiagm may enhance survival prediction in PDAC, warranting large-scale studies to confirm its applicability in clinical practice.
Collapse
Affiliation(s)
- Yeon-Koo Kang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seunggyun Ha
- Division of Nuclear Medicine, Department of Radiology, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, Seoul, Republic of Korea
| | - Ji Bong Jeong
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| | - So Won Oh
- Department of Nuclear Medicine, Seoul Metropolitan Government - Seoul National University Boramae Medical Center, Seoul National University College of Medicine, 20 Boramae-ro 5-gil, Dongjak-Gu, Seoul, Republic of Korea.
| |
Collapse
|
3
|
Li J, Du J, Li Y, Meng M, Hang J, Shi H. A nomogram based on CT texture features to predict the response of patients with advanced pancreatic cancer treated with chemotherapy. BMC Gastroenterol 2023; 23:274. [PMID: 37563572 PMCID: PMC10416463 DOI: 10.1186/s12876-023-02902-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE This study aimed to evaluate the predictive value of computed tomography (CT) texture features in the treatment response of patients with advanced pancreatic cancer (APC) receiving palliative chemotherapy. METHODS This study enrolled 84 patients with APC treated with first-line chemotherapy and conducted texture analysis on primary pancreatic tumors. 59 patients and 25 were randomly assigned to the training and validation cohorts at a ratio of 7:3. The treatment response to chemotherapy was evaluated according to the Response Evaluation Criteria in Solid Tumors (RECIST1.1). The patients were divided into progressive and non-progressive groups. The least absolute shrinkage selection operator (LASSO) was applied for feature selection in the training cohort and a radiomics signature (RS) was calculated. A nomogram was developed based on a multivariate logistic regression model incorporating the RS and carbohydrate antigen 19-9 (CA19-9), and was internally validated using the C-index and calibration plot. We performed the decision curve analysis (DCA) and clinical impact curve analysis to reflect the clinical utility of the nomogram. The nomogram was further externally confirmed in the validation cohort. RESULTS The multivariate logistic regression analysis indicated that the RS and CA19-9 were independent predictors (P < 0.05), and a trend was found for chemotherapy between progressive and non-progressive groups. The nomogram incorporating RS, CA19-9 and chemotherapy showed favorable discriminative ability in the training (C-index = 0.802) and validation (C-index = 0.920) cohorts. The nomogram demonstrated favorable clinical utility. CONCLUSION The RS of significant texture features was significantly associated with the early treatment effect of patients with APC treated with chemotherapy. Based on the RS, CA19-9 and chemotherapy, the nomogram provided a promising way to predict chemotherapeutic effects for APC patients.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Jiadi Du
- Department of Computer Science, Missouri University of Science and Technology, Rolla, MO, U.S
| | - Yuying Li
- Graduate College, Dalian Medical University, Dalian, China
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Mingzhu Meng
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China
| | - Junjie Hang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, 518116, Shenzhen, China.
- Department of Oncology, the Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou Medical Center, Changzhou, China.
| | - Haifeng Shi
- Department of Radiology, Changzhou Second People's Hospital, Changzhou, China.
| |
Collapse
|
4
|
Zhao Y, Liu Y, Jia Y, Wang X, He J, Zhen S, Wang J, Liu L. Fibroblast activation protein in the tumor microenvironment predicts outcomes of PD-1 blockade therapy in advanced non-small cell lung cancer. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04250-4. [PMID: 35951090 DOI: 10.1007/s00432-022-04250-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE The identification of robust predictive biomarkers of the response to programmed cell death-1 (PD-1) blockade remains a critical concern. Here, we investigated on fibroblast activation protein (FAP) as a microenvironment-derived biomarker of clinical outcomes of PD-1 blockade therapy, and the correlation between FAP expression and T cell infiltration in advanced non-small cell lung cancer (NSCLC). METHODS A total of 135 patients with advanced NSCLC who received PD-1 blockade therapy were retrospectively analyzed. The potential associations among FAP expression, CD3 + T cell and CD8 + T cell infiltration, and clinical outcomes of immunotherapy were validated by immunohistochemistry, bioinformatic analyses, and statistical measurements. RESULTS FAP was widely expressed in advanced NSCLC tissues. FAP was correlated with decreased density of CD8 + T cells (Spearman's rho - 0.32, p < 0.001) and immunosuppressive tumor microenvironment (TME) status. No correlations were detected between FAP and PD-L1 expression or with the density of CD3 + T cells. The patients with higher expression of FAP showed worse response rate (16.4% vs. 38.7%, p < 0.001) and worse progression-free survival (HR = 2.56, 95% CI 1.69-3.87, p < 0.001). In addition, FAP contributed to shortened overall survival in subgroups of the patients with squamous cell lung cancer (p = 0.020), PD-1 blockade monotherapy (p = 0.017), and first-line therapy (p = 0.028). CONCLUSION FAP is a potential predictive biomarker of resistance to PD-1 blockade. Further investigation is warranted to identify a strategy for targeting FAP to alleviate the immunosuppressive TME and broaden the clinical effectiveness of PD-1 blockade therapy.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China.,Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Xiaoxiao Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Jiankun He
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, China
| | - Shuman Zhen
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050035, China. .,Cancer Research Institute of Hebei Province, Shijiazhuang, 050011, China. .,China International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
5
|
A systematic review of prognosis predictive role of radiomics in pancreatic cancer: heterogeneity markers or statistical tricks? Eur Radiol 2022; 32:8443-8452. [PMID: 35904618 DOI: 10.1007/s00330-022-08922-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES We aimed to systematically evaluate the prognostic prediction accuracy of radiomics features extracted from pre-treatment imaging in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS Radiomics literature on overall survival (OS) prediction of PDAC were all included in this systematic review. A further meta-analysis was performed on the effect size of first-order entropy. Methodological quality and risk of bias of the included studies were assessed by the radiomics quality score (RQS) and prediction model risk of bias assessment tool (PROBAST). RESULTS Twenty-three studies were finally identified in this review. Two (8.7%) studies compared prognosis prediction ability between radiomics model and TNM staging model by C-index, and both showed a better performance of the radiomics. Twenty-one (91.3%) studies reported significant predictive values of radiomics features. Nine (39.1%) studies were included in the meta-analysis, and it showed a significant correlation between first-order entropy and OS (HR 1.66, 95%CI 1.18-2.34). RQS assessment revealed validation was only performed in 5 (21.7%) studies on internal datasets and 2 (8.7%) studies on external datasets. PROBAST showed that 22 (95.7%) studies have a high risk of bias in participants because of the retrospective study design. CONCLUSION First-order entropy was significantly associated with OS and might improve the accuracy of PDAC prognosis prediction. Existing studies were poorly validated, and it should be noted in future studies. Modification of PROBAST for radiomics studies is necessary since the strict requirements of prospective study design may not be applicable to the demand for a large sample size in the model construction stage. KEY POINTS • Radiomics based on the primary lesion holds great potential for prognosis prediction. First-order entropy was significantly associated with the overall survival of PDAC and might improve the accuracy of current PDAC prognosis prediction. • We strongly recommend that at least an internal validation should be conducted in any radiomics study. Attention should be paid to the complex relationships between radiomics features. • Due to the close relationship between radiomics and big data, the strict requirement of prospective study design in PROABST may not be appropriate for radiomics studies. A balance between study types and sample sizes for radiomics studies needs to be found in the model construction stage.
Collapse
|
6
|
Preuss K, Thach N, Liang X, Baine M, Chen J, Zhang C, Du H, Yu H, Lin C, Hollingsworth MA, Zheng D. Using Quantitative Imaging for Personalized Medicine in Pancreatic Cancer: A Review of Radiomics and Deep Learning Applications. Cancers (Basel) 2022; 14:cancers14071654. [PMID: 35406426 PMCID: PMC8997008 DOI: 10.3390/cancers14071654] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/12/2022] Open
Abstract
Simple Summary With a five-year survival rate of only 3% for the majority of patients, pancreatic cancer is a global healthcare challenge. Radiomics and deep learning, two novel quantitative imaging methods that treat medical images as minable data instead of just pictures, have shown promise in advancing personalized management of pancreatic cancer through diagnosing precursor diseases, early detection, accurate diagnosis, and treatment personalization. Radiomics and deep learning methods aim to collect hidden information in medical images that is missed by conventional radiology practices through expanding the data search and comparing information across different patients. Both methods have been studied and applied in pancreatic cancer. In this review, we focus on the current progress of these two methods in pancreatic cancer and provide a comprehensive narrative review on the topic. With better regulation, enhanced workflow, and larger prospective patient datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through personalized precision medicine. Abstract As the most lethal major cancer, pancreatic cancer is a global healthcare challenge. Personalized medicine utilizing cutting-edge multi-omics data holds potential for major breakthroughs in tackling this critical problem. Radiomics and deep learning, two trendy quantitative imaging methods that take advantage of data science and modern medical imaging, have shown increasing promise in advancing the precision management of pancreatic cancer via diagnosing of precursor diseases, early detection, accurate diagnosis, and treatment personalization and optimization. Radiomics employs manually-crafted features, while deep learning applies computer-generated automatic features. These two methods aim to mine hidden information in medical images that is missed by conventional radiology and gain insights by systematically comparing the quantitative image information across different patients in order to characterize unique imaging phenotypes. Both methods have been studied and applied in various pancreatic cancer clinical applications. In this review, we begin with an introduction to the clinical problems and the technology. After providing technical overviews of the two methods, this review focuses on the current progress of clinical applications in precancerous lesion diagnosis, pancreatic cancer detection and diagnosis, prognosis prediction, treatment stratification, and radiogenomics. The limitations of current studies and methods are discussed, along with future directions. With better standardization and optimization of the workflow from image acquisition to analysis and with larger and especially prospective high-quality datasets, radiomics and deep learning methods could show real hope in the battle against pancreatic cancer through big data-based high-precision personalization.
Collapse
Affiliation(s)
- Kiersten Preuss
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Nutrition and Health Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA
| | - Nate Thach
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Xiaoying Liang
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Michael Baine
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Justin Chen
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Naperville North High School, Naperville, IL 60563, USA
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Huijing Du
- Department of Mathematics, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Hongfeng Yu
- Department of Computer Science, University of Nebraska Lincoln, Lincoln, NE 68588, USA;
| | - Chi Lin
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
| | - Michael A. Hollingsworth
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Dandan Zheng
- Department of Radiation Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA; (K.P.); (N.T.); (M.B.); (J.C.); (C.L.)
- Department of Radiation Oncology, University of Rochester, Rochester, NY 14626, USA
- Correspondence: ; Tel.: +1-(585)-276-3255
| |
Collapse
|