1
|
Wang Z, Zhu L, Wang Y, Han X, Xu Q, Dai M. Looking at or beyond the tumor - a systematic review and meta-analysis of quantitative imaging biomarkers predicting pancreatic cancer prognosis. Abdom Radiol (NY) 2025:10.1007/s00261-025-04919-7. [PMID: 40195140 DOI: 10.1007/s00261-025-04919-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/15/2025] [Accepted: 03/26/2025] [Indexed: 04/09/2025]
Abstract
OBJECTIVES To evaluate the prognostic value of quantitative imaging biomarkers derived from computed tomography (CT) and magnetic resonance imaging (MRI) for pancreatic cancer (PC), with a particular focus on body composition parameters beyond the traditional intrinsic features of the tumor. METHODS PubMed, EMBASE, and Cochrane Library databases were searched for articles on quantitative imaging biomarkers obtained from CT or MRI in predicting PC prognosis published between January 2014 and August 2024. The Newcastle-Ottawa scale was used to assess the quality of the included studies. Survival outcomes, such as overall survival (OS) and recurrence-free survival (RFS), were evaluated. The pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated using a random-effects model. In case of high heterogeneity, subgroup analyses and sensitivity analyses were performed to identify potential sources of heterogeneity among the studies. RESULTS We performed a meta-analysis of ten imaging biomarkers investigated in 43 included studies. Larger tumor size, lower skeletal muscle radiodensity, lower skeletal muscle index (SMI), presence of sarcopenic obesity, lower psoas muscle index (PMI), higher visceral to subcutaneous adipose tissue area ratio, and lower visceral adipose tissue index were associated with significantly worse OS. In particular, lower SMI and lower PMI had relatively high HRs (1.65 for SMI, 95% CI 1.39-1.96, and 2.20 for PMI, 95% CI 1.74-2.78). Patients with lower SMI exhibited poorer RFS (HR 1.78, 95% CI 1.46-2.18). Subgroup analyses identified the origin region of the study and intervention type as potential factors of heterogeneity for SMI in predicting OS. CONCLUSIONS Imaging biomarkers indicating body composition at PC diagnosis may play an important role in predicting patient prognosis. Further prospective multi-center studies with large sample sizes are needed for validation and translation into clinical practice.
Collapse
Affiliation(s)
- Zihe Wang
- School of Medicine, Anhui Medical University, Hefei, China
| | - Liang Zhu
- Department of Radiology, Peking Union Medical College Hospital, Beijing, China.
| | - Yitan Wang
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut, USA
| | - Xianlin Han
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
2
|
Boubaddi M, Rossi J, Marichez A, Marty M, Amintas S, Laurent C, Dabernat S. Preoperative Prognostic Factors in Resectable Pancreatic Cancer: State of the Art and Prospects. Ann Surg Oncol 2025:10.1245/s10434-025-17062-w. [PMID: 40095311 DOI: 10.1245/s10434-025-17062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/09/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Only 15% to 20% of patients with pancreatic ductal adenocarcinoma (PDAC) have access to surgical resection, which represents the only chance of curative treatment. Current resection classifications are almost exclusively anatomic and do not correlate sufficiently with patient survival. It is essential to develop preoperative prognostic factors to distinguish patients at high risk of early postoperative recurrence from those who will have prolonged survival after surgery. In some cases, PDACs may present biomolecular differences reflecting their aggressiveness that are not yet assessable by the current clinical-biologic assessment. This study aimed to assess the preoperative prognostic factors that are already available and the future perspectives being developed. METHOD This study reviewed the literature using the PubMed public database for preoperative prognostic factors for resectable PDAC. CONCLUSION Validated preoperative prognostic factors, whether clinical, biologic, radiologic, or histologic, are very important in anticipating the course of each patient's disease. The identification of potential new prognostic biomarkers such as genomic, transcriptomic, and proteomic analyses and the dosage of circulating tumor DNA are very serious avenues to be developed, but the extraction and analysis techniques as well as the interpretation of their results need to be standardized in prospective studies.
Collapse
Affiliation(s)
- Mehdi Boubaddi
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France.
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France.
- Hepatobiliary and Pancreatic Surgery Department, Bordeaux University Hospital, Bordeaux, France.
| | - Julia Rossi
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Arthur Marichez
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Marion Marty
- Tumor Biology and Tumor Bank Laboratory, CHU Bordeaux, Bordeaux, France
| | - Samuel Amintas
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Christophe Laurent
- Colorectal Unit, Department of Digestive Surgery, Bordeaux University Hospital, Bordeaux, France
| | - Sandrine Dabernat
- BRIC (BoRdeaux Institute of onCology), UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| |
Collapse
|
3
|
Mohamed SA, Barlemann A, Steinle V, Nonnenmacher T, Güttlein M, Hackert T, Loos M, Gaida MM, Kauczor HU, Klauss M, Mayer P. Performance of different CT enhancement quantification methods as predictors of pancreatic cancer recurrence after upfront surgery. Sci Rep 2024; 14:19783. [PMID: 39187515 PMCID: PMC11347575 DOI: 10.1038/s41598-024-70441-3] [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: 03/07/2024] [Accepted: 08/16/2024] [Indexed: 08/28/2024] Open
Abstract
The prognosis of pancreatic cancer (PDAC) after tumor resection remains poor, mostly due to a high but variable risk of recurrence. A promising tool for improved prognostication is the quantification of CT tumor enhancement. For this, various enhancement formulas have been used in previous studies. However, a systematic comparison of these formulas is lacking. In the present study, we applied twenty-three previously published CT enhancement formulas to our cohort of 92 PDAC patients who underwent upfront surgery. We identified seven formulas that could reliably predict tumor recurrence. Using these formulas, weak tumor enhancement was associated with tumor recurrence at one and two years after surgery (p ≤ 0.030). Enhancement was inversely associated with adverse clinicopathological features. Low enhancement values were predictive of a high recurrence risk (Hazard Ratio ≥ 1.659, p ≤ 0.028, Cox regression) and a short time to recurrence (TTR) (p ≤ 0.027, log-rank test). Some formulas were independent predictors of TTR in multivariate models. Strikingly, almost all of the best-performing formulas measure solely tumor tissue, suggesting that normalization to non-tumor structures might be unnecessary. Among the top performers were also the absolute arterial/portal venous tumor attenuation values. These can be easily implemented in clinical practice for better recurrence prediction, thus potentially improving patient management.
Collapse
Affiliation(s)
- Sherif A Mohamed
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
- Department of Neuroradiology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Alina Barlemann
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Verena Steinle
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Tobias Nonnenmacher
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Michelle Güttlein
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Loos
- Clinic of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Matthias M Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- TRON, Translational Oncology at the University Medical Center, JGU-Mainz, Mainz, Germany
| | - Hans-Ulrich Kauczor
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Miriam Klauss
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Philipp Mayer
- Clinic for Diagnostic and Interventional Radiology, Heidelberg University Hospital, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany.
| |
Collapse
|
4
|
Yamaguchi T, Sofue K, Ueshima E, Sugiyama N, Yabe S, Ueno Y, Masuda A, Toyama H, Kodama T, Komatsu M, Hori M, Murakami T. Rim Enhancement on Contrast-Enhanced CT as a Predictor of Prognosis in Patients with Pancreatic Ductal Adenocarcinoma. Diagnostics (Basel) 2024; 14:782. [PMID: 38667428 PMCID: PMC11048909 DOI: 10.3390/diagnostics14080782] [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: 03/06/2024] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/28/2024] Open
Abstract
This study investigated the utility of imaging features, such as rim enhancement on contrast-enhanced CT (CECT), in predicting the prognosis of pancreatic ductal adenocarcinoma (PDAC). This retrospective study included 158 patients (84 men; mean age, 68 years) with pathologically confirmed PDAC. The following imaging features were evaluated on CECT by two radiologists: tumor size, tumor attenuation, and the presence of rim enhancement. Cox proportional hazards analysis was performed to identify the imaging and clinicopathological features for predicting disease-free survival (DFS) and overall survival (OS). Pathological features were compared with the presence of rim enhancement. Among the 158 patients, 106 (67%) underwent curative surgery (surgery group) and 52 (33%) received conservative treatment (non-surgery group). Rim enhancement was observed more frequently in the non-surgery group than in the surgery group (44% vs. 20%; p < 0.001). Rim enhancement showed significant associations with shorter DFS and OS in the surgery group (hazard ratios (HRs), 3.03 and 2.99; p < 0.001 and p = 0.003, respectively), whereas tumor size showed significant associations with shorter OS (HR per 1 mm increase, 1.08; p < 0.001). PDACs with rim enhancement showed significant associations with higher histological tumor grades (p < 0.001). PDAC with rim enhancement on CECT could predict poorer prognosis and more aggressive tumor grades.
Collapse
Affiliation(s)
- Takeru Yamaguchi
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Eisuke Ueshima
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Naoki Sugiyama
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Shinji Yabe
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Yoshiko Ueno
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Atsuhiro Masuda
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Hirochika Toyama
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takayuki Kodama
- Department of Pathology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masato Komatsu
- Department of Pathology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Masatoshi Hori
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-cho, Chuo-ku, Kobe 650-0017, Japan
| |
Collapse
|
5
|
Saleh M, Virarkar M, Mahmoud HS, Wong VK, Gonzalez Baerga CI, Parikh M, Elsherif SB, Bhosale PR. Radiomics analysis with three-dimensional and two-dimensional segmentation to predict survival outcomes in pancreatic cancer. World J Radiol 2023; 15:304-314. [PMID: 38058604 PMCID: PMC10696186 DOI: 10.4329/wjr.v15.i11.304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 10/23/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Radiomics can assess prognostic factors in several types of tumors, but considering its prognostic ability in pancreatic cancer has been lacking. AIM To evaluate the performance of two different radiomics software in assessing survival outcomes in pancreatic cancer patients. METHODS We retrospectively reviewed pretreatment contrast-enhanced dual-energy computed tomography images from 48 patients with biopsy-confirmed pancreatic ductal adenocarcinoma who later underwent neoadjuvant chemoradiation and surgery. Tumors were segmented using TexRad software for 2-dimensional (2D) analysis and MIM software for 3D analysis, followed by radiomic feature extraction. Cox proportional hazard modeling correlated texture features with overall survival (OS) and progression-free survival (PFS). Cox regression was used to detect differences in OS related to pretreatment tumor size and residual tumor following treatment. The Wilcoxon test was used to show the relationship between tumor volume and the percent of residual tumor. Kaplan-Meier analysis was used to compare survival in patients with different tumor densities in Hounsfield units for both 2D and 3D analysis. RESULTS 3D analysis showed that higher mean tumor density [hazard ratio (HR) = 0.971, P = 0.041)] and higher median tumor density (HR = 0.970, P = 0.037) correlated with better OS. 2D analysis showed that higher mean tumor density (HR = 0.963, P = 0.014) and higher mean positive pixels (HR = 0.962, P = 0.014) correlated with better OS; higher skewness (HR = 3.067, P = 0.008) and higher kurtosis (HR = 1.176, P = 0.029) correlated with worse OS. Higher entropy correlated with better PFS (HR = 0.056, P = 0.036). Models determined that patients with increased tumor size greater than 1.35 cm were likely to have a higher percentage of residual tumors of over 10%. CONCLUSION Several radiomics features can be used as prognostic tools for pancreatic cancer. However, results vary between 2D and 3D analyses. Mean tumor density was the only variable that could reliably predict OS, irrespective of the analysis used.
Collapse
Affiliation(s)
- Mohammed Saleh
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Mayur Virarkar
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Hagar S Mahmoud
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Vincenzo K Wong
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Carlos Ignacio Gonzalez Baerga
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Miti Parikh
- Keck School of Medicine, University of South California, Los Angeles, CA 90033, United States
| | - Sherif B Elsherif
- Department of Diagnostic Radiology, The University of Florida College of Medicine, Jacksonville, FL 32209, United States
| | - Priya R Bhosale
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
6
|
Shao Y, Dang Y, Cheng Y, Gui Y, Chen X, Chen T, Zeng Y, Tan L, Zhang J, Xiao M, Yan X, Lv K, Zhou Z. Predicting the Efficacy of Neoadjuvant Chemotherapy for Pancreatic Cancer Using Deep Learning of Contrast-Enhanced Ultrasound Videos. Diagnostics (Basel) 2023; 13:2183. [PMID: 37443577 DOI: 10.3390/diagnostics13132183] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Contrast-enhanced ultrasound (CEUS) is a promising imaging modality in predicting the efficacy of neoadjuvant chemotherapy for pancreatic cancer, a tumor with high mortality. In this study, we proposed a deep-learning-based strategy for analyzing CEUS videos to predict the prognosis of pancreatic cancer neoadjuvant chemotherapy. Pre-trained convolutional neural network (CNN) models were used for binary classification of the chemotherapy as effective or ineffective, with CEUS videos collected before chemotherapy as the model input, and with the efficacy after chemotherapy as the reference standard. We proposed two deep learning models. The first CNN model used videos of ultrasound (US) and CEUS (US+CEUS), while the second CNN model only used videos of selected regions of interest (ROIs) within CEUS (CEUS-ROI). A total of 38 patients with strict restriction of clinical factors were enrolled, with 76 original CEUS videos collected. After data augmentation, 760 and 720 videos were included for the two CNN models, respectively. Seventy-six-fold and 72-fold cross-validations were performed to validate the classification performance of the two CNN models. The areas under the curve were 0.892 and 0.908 for the two models. The accuracy, recall, precision and F1 score were 0.829, 0.759, 0.786, and 0.772 for the first model. Those were 0.864, 0.930, 0.866, and 0.897 for the second model. A total of 38.2% and 40.3% of the original videos could be clearly distinguished by the deep learning models when the naked eye made an inaccurate classification. This study is the first to demonstrate the feasibility and potential of deep learning models based on pre-chemotherapy CEUS videos in predicting the efficacy of neoadjuvant chemotherapy for pancreas cancer.
Collapse
Affiliation(s)
- Yuming Shao
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yingnan Dang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yang Gui
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xueqi Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Tianjiao Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yan Zeng
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| | - Li Tan
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Mengsu Xiao
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaoyi Yan
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ke Lv
- Department of Ultrasound, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhuhuang Zhou
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
7
|
He M, Wang X, Xu J, Li J, Chang X, Zins M, Jin Z, Xue H. Diffuse Involvement of Pancreas is not Always Autoimmune Pancreatitis. Acad Radiol 2022; 29:1523-1531. [PMID: 35279380 DOI: 10.1016/j.acra.2022.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/09/2022] [Accepted: 01/16/2022] [Indexed: 02/07/2023]
Abstract
RATIONALE AND OBJECTIVES To determine the prevalence of diffuse involvement of pancreas and to identify the findings of malignancies using enhancement computed tomography (CT). MATERIALS AND METHODS A total of 1,0249 patients performed enhancement CT in our hospital over 62 months were investigated and the final study cohort includes 245 patients (170 males, 75 females; mean age, 56.94 ± 12.17 years). The reference standard is the final clinical/pathological diagnosis. The lesion-to-aorta enhancement ratio (LAR) on the pancreatic arterial phase, portal phase and delayed phase (DP) and the traditional CT findings were evaluated. Intergroup comparisons between malignancies and non-malignancies lesions were performed. Univariate and multivariate analyses were conducted to identify findings predicting malignancies. RESULTS The prevalence of malignancy was 45.3% (111/245) of diffuse enlargement of pancreas. All benign lesions were autoimmune pancreatitis 54.7% (n = 134). The most common malignant lesion was pancreatic ductal adenocarcinoma (n = 88, 35.9%). Other rare lesions with malignant potential included pancreatic neuroendocrine tumor (n = 11, 4.5%), lymphoma (n = 4, 1.6%), metastasis (n = 4, 1.6%), solid pseudopapillary neoplasm (n = 3, 1.2%) and acinar cell carcinoma (n = 1, 0.4%). Residual normal pancreas parenchyma, heterogeneity, short axis (cut-off value, 3.15 cm) and LARDP (cut-off value, 0.75) were independent predictors of malignancies. When the above predictors were combined, a sensitivity of 94.2%, a specificity of 90.8% were attained. CONCLUSION Diffuse involvement of the pancreas is rare and is not a specific sign of autoimmune pancreatitis, and it is associated with a wide spectrum of malignant conditions. Dynamic enhancement CT is helpful to identifying malignancies.
Collapse
Affiliation(s)
- Ming He
- Department of Radiology, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, People's Republic of China; Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China
| | - Xiheng Wang
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China
| | - Jin Xu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China
| | - Juan Li
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Marc Zins
- Department of Rathology, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Zhengyu Jin
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China
| | - Huadan Xue
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan No.1, Dongcheng District, Beijing 100703, China.
| |
Collapse
|
8
|
Barat M, Marchese U, Pellat A, Dohan A, Coriat R, Hoeffel C, Fishman EK, Cassinotto C, Chu L, Soyer P. Imaging of Pancreatic Ductal Adenocarcinoma: An Update on Recent Advances. Can Assoc Radiol J 2022; 74:351-361. [PMID: 36065572 DOI: 10.1177/08465371221124927] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pancreatic ductal carcinoma (PDAC) is one of the leading causes of cancer-related death worldwide. Computed tomography (CT) remains the primary imaging modality for diagnosis of PDAC. However, CT has limitations for early pancreatic tumor detection and tumor characterization so that it is currently challenged by magnetic resonance imaging. More recently, a particular attention has been given to radiomics for the characterization of pancreatic lesions using extraction and analysis of quantitative imaging features. In addition, radiomics has currently many applications that are developed in conjunction with artificial intelligence (AI) with the aim of better characterizing pancreatic lesions and providing a more precise assessment of tumor burden. This review article sums up recent advances in imaging of PDAC in the field of image/data acquisition, tumor detection, tumor characterization, treatment response evaluation, and preoperative planning. In addition, current applications of radiomics and AI in the field of PDAC are discussed.
Collapse
Affiliation(s)
- Maxime Barat
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| | - Ugo Marchese
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Digestive, Hepatobiliary and Pancreatic Surgery, 26935Hopital Cochin, AP-HP, Paris, France
| | - Anna Pellat
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Gastroenterology, 26935Hopital Cochin, AP-HP, Paris, France
| | - Anthony Dohan
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| | - Romain Coriat
- Université Paris Cité, Faculté de Médecine, 555089Paris, France.,Department of Gastroenterology, 26935Hopital Cochin, AP-HP, Paris, France
| | | | - Elliot K Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, 1466Johns Hopkins University, Baltimore, MD, USA
| | - Christophe Cassinotto
- Department of Radiology, CHU Montpellier, 27037University of Montpellier, Saint-Éloi Hospital, Montpellier, France
| | - Linda Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, 1466Johns Hopkins University, Baltimore, MD, USA
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris543341, Paris, France.,Université Paris Cité, Faculté de Médecine, 555089Paris, France
| |
Collapse
|
9
|
Tian N, Wu D, Zhu L, Zeng M, Li J, Wang X. A predictive model for recurrence after upfront surgery in patients with resectable pancreatic ductal adenocarcinoma (PDAC) by using preoperative clinical data and CT characteristics. BMC Med Imaging 2022; 22:116. [PMID: 35786426 PMCID: PMC9252003 DOI: 10.1186/s12880-022-00823-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The overall survival for patients with resectable PDAC following curative surgical resection hasn't been improved significantly, as a considerable proportion of patients develop recurrence within a year. The purpose of this study was to develop and validate a predictive model to assess recurrence risk in patients with PDAC after upfront surgery by using preoperative clinical data and CT characteristics. METHODS The predictive model was developed based on a retrospective set of 141 pancreatic cancer patients after surgery. A separate set of 77 patients was used to validate model. Between January 2017 and December 2019, all patients underwent multidetector pancreatic CT and upfront surgery. Univariable and multivariate Cox regression was used to determine the risk factors related to recurrence and then establish a nomogram to estimate the 1-year recurrence probability. The Harrell C-index was employed in evaluating the discrimination and calibration of the model. RESULTS A total of 218 patients in this retrospective cohort. A recurrence model in nomogram form was developed with predictors including tumor size (hazard ratio [HR], 1.277; 95% CI 1.098, 1.495; P = 0.002), tumor density in the portal vein phase (HR, 0.598; 95% CI 0.424, 0.844; P = 0.003), peripancreatic infiltration (HR, 4.151; 95% CI 2.077, 8.298; P < 0.001), suspicious metastatic lymph node (HR, 2.561; 95% CI 1.653, 3.967; P < 0.001), Neutrophils/Lymphocytes ratio (HR, 1.111; 95% CI 1.016, 1.215; P = 0.020). The predictive nomogram had good discrimination capability with these predictors with an area under curve at 1 year of 0.84 (95%CI 0.77, 0.91) in the development set and 0.82 (95% CI 0.72, 0.92) and 0.84 (95% CI 0.74, 0.94) in the validation set for two radiologists reading respectively. CONCLUSIONS The model developed based on preoperative clinical data and CT characteristics of resectable pancreatic ductal adenocarcinoma patients, which can helpfully estimate the recurrence-free survival. It may be a useful tool for clinician to select optimal candidates for upfront surgery or neoadjuvant therapy.
Collapse
Affiliation(s)
- Ningzi Tian
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Dong Wu
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Lei Zhu
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Mengsu Zeng
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
- Department of Radiology, Zhongshan Hospital, Fudan University, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Jianke Li
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China
| | - Xiaolin Wang
- Shanghai Institute of Medical Imaging, No. 180 Fenglin Road, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
10
|
Gao JF, Pan Y, Lin XC, Lu FC, Qiu DS, Liu JJ, Huang HG. Prognostic value of preoperative enhanced computed tomography as a quantitative imaging biomarker in pancreatic cancer. World J Gastroenterol 2022; 28:2468-2481. [PMID: 35979266 PMCID: PMC9258279 DOI: 10.3748/wjg.v28.i22.2468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/31/2021] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with high mortality and short survival time. Computed tomography (CT) plays an important role in the diagnosis, staging and treatment of pancreatic tumour. Pancreatic cancer generally shows a low enhancement pattern compared with normal pancreatic tissue.
AIM To analyse whether preoperative enhanced CT could be used to predict postoperative overall survival in patients with PDAC.
METHODS Sixty-seven patients with PDAC undergoing pancreatic resection were enrolled retrospectively. All patients underwent preoperative unenhanced and enhanced CT examination, the CT values of which were measured. The ratio of the preoperative CT value increase from the nonenhancement phase to the portal venous phase between pancreatic tumour and normal pancreatic tissue was calculated. The cut-off value of ratios was obtained by the receiver operating characteristic (ROC) curve of the tumour relative enhancement ratio (TRER), according to which patients were divided into low- and high-enhancement groups. Univariate and multivariate analyses were performed using Cox regression based on TRER grouping. Finally, the correlation between TRER and clinicopathological characteristics was analysed.
RESULTS The area under the curve of the ROC curve was 0.768 (P < 0.05), and the cut-off value of the ROC curve was calculated as 0.7. TRER ≤ 0.7 was defined as the low-enhancement group, and TRER > 0.7 was defined as the high-enhancement group. According to the TRER grouping, the Kaplan-Meier survival curve analysis results showed that the median survival (10.0 mo) with TRER ≤ 0.7 was significantly shorter than that (22.0 mo) with TRER > 0.7 (P < 0.05). In the univariate and multivariate analyses, the prognosis of patients with TRER ≤ 0.7 was significantly worse than that of patients with TRER > 0.7 (P < 0.05). Our results demonstrated that patients in the low TRER group were more likely to have higher American Joint Committee on Cancer stage, tumour stage and lymph node stage (all P < 0.05), and TRER was significantly negatively correlated with tumour size (P < 0.05).
CONCLUSION TRER ≤ 0.7 in patients with PDAC may represent a tumour with higher clinical stage and result in a shorter overall survival.
Collapse
Affiliation(s)
- Jian-Feng Gao
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Xian-Chao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Feng-Chun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| | - Ding-Shen Qiu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - Jun-Jun Liu
- Department of Radiology, The Hospital of Changle, Fuzhou 350200, Fujian Province, China
| | - He-Guang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
11
|
Abstract
The basic pancreatic lesions include location, size, shape, number, capsule, calcification/calculi, hemorrhage, cystic degeneration, fibrosis, pancreatic duct alterations, and microvessel. One or more basic lesions form a kind of pancreatic disease. As recognizing the characteristic imaging features of pancreatic basic lesions and their relationships with pathology aids in differentiating the variety of pancreatic diseases. The purpose of this study is to review the pathological and imaging features of the basic pancreatic lesions.
Collapse
|
12
|
Zhou T, Tan L, Gui Y, Zhang J, Chen X, Dai M, Xiao M, Zhang Q, Chang X, Xu Q, Bai C, Cheng Y, Xu Q, Wang X, Meng H, Jia W, Lv K, Jiang Y. Correlation Between Enhancement Patterns on Transabdominal Ultrasound and Survival for Pancreatic Ductal Adenocarcinoma. Cancer Manag Res 2021; 13:6823-6832. [PMID: 34512022 PMCID: PMC8418377 DOI: 10.2147/cmar.s307079] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/12/2021] [Indexed: 12/30/2022] Open
Abstract
Objective Investigate the CEUS enhancement patterns of PDAC and analyse correlations between the CEUS enhancement pattern and both the degree of tumour tissue differentiation and overall survival (OS). Methods The study included 56 patients with locally advanced PDAC, performed conventional ultrasound and CEUS, and analysed characteristics of the CEUS enhancement patterns. In addition, clinical data, such as serum level of CA19-9, TNM stage were collected, and patients’ survival times were followed up. TICs of dynamic CEUS images were acquired using image processing software to obtain the peak, TP, sharpness, and AUC. Correlations of the CEUS enhancement patterns of PDAC with the degree of differentiation of tumour tissue and OS were quantitatively analysed, as were the correlations of the TIC parameters and CEUS enhancement patterns with OS. Results Enhancement in the arterial phase included iso-enhancement (30.3%) and hypo-enhancement (69.6%), and was not significantly correlated with sex, age at disease onset, or lesion size. Also was not significantly correlated with tumour tissue differentiation. Clear survival times were obtained for 50 patients during follow-up, and the median survival time was significantly longer for the patients with iso-enhancement than hypo-enhancement. Among the TIC parameters, peaktumour, sharpnesstumour, AEsharpness, and REsharpness differed significantly between the group with iso-enhancement and hypo-enhancement (p < 0.05). Conclusion The CEUS enhancement patterns of PDAC in the arterial phase include iso-enhancement and hypo-enhancement. Enhancement pattern was not significantly correlated with the degree of differentiation of tumour tissue, but patient survival time differed significantly between the two enhancement patterns, with longer survival for patients with iso-enhancement.
Collapse
Affiliation(s)
- Tongtong Zhou
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Department of Ultrasound, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Li Tan
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yang Gui
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Jing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xueqi Chen
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Menghua Dai
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Mengsu Xiao
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qing Zhang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qun Xu
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, People's Republic of China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yuejuan Cheng
- Department of Medical Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Xue Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People's Republic of China
| | - Hua Meng
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Wanying Jia
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Ke Lv
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| | - Yuxin Jiang
- Department of Ultrasound, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
13
|
Shantarevich MY, Karmazanovsky GG, Egorkina AB, Kurochkina AI, Kriger AG, Kalinin DV, Stashkiv VI. [Computed tomography in determining the differentiation of ductal adenocarcinoma of pancreatic head]. Khirurgiia (Mosk) 2021:11-19. [PMID: 33710821 DOI: 10.17116/hirurgia202103111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To evaluate the features of «hypervascular rim», tumor dimensions and density as prognostic factors of differentiation of pancreatic head adenocarcinoma. MATERIAL AND METHODS Pancreatoduodenectomy was performed in 311 patients with pancreatic head adenocarcinoma for the period 2013-2019. A retrospective study included 81 patients who met the following criteria: available data of morphological and immunohistological examination indicating tumor grade from Grade 1 to Grade 3, as well as available preoperative CT images in four phases (native, arterial, portal and delayed). Tumor dimensions, density of the pancreas, adenocarcinoma and abdominal aorta by the phases of contrast enhancement were analyzed in all patients. Moreover, we estimated coefficient of relative enhancement change. Perifocal hypervascular enhancement was assessed in arterial and portal phases. Contrast-enhanced MRI was performed in 15 out of 81 patients. MR images were analyzed regarding a hypervascular rim, and the last one was compared with CT images. RESULTS There was no significant difference in density values between different tumor grades. Coefficient of relative enhancement change >1 was observed in 63.64% of highly-differentiated tumors. REC ≤1 was found in 85.11% of tumors grade 2 and 82.6% of tumors grade 3 (p=0.005). According to Chi-square test, there was a correlation between tumor differentiation and hypervascular rim (p=0.03). Moderate and low differentiation was observed in 96.42% of tumors with perifocal enhancement. Hypervascular rim was absent in 81.82% of tumors grade 1. Adenocarcinoma grade 2 was found in 85.71% of cases with unclear perifocal enhancement. CONCLUSION Preoperative contrast-enhanced CT is valuable to assume the tumor grade in patients with pancreatic head adenocarcinoma due to assessment of hypervascular rim and REC.
Collapse
Affiliation(s)
- M Yu Shantarevich
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia
| | - G G Karmazanovsky
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia.,Pirogov Russian National Research Medical University, Moscow, Russia
| | - A B Egorkina
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia
| | - A I Kurochkina
- Federal Research Institute for Health Organization and Informatics, Moscow, Russia
| | - A G Kriger
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia
| | - D V Kalinin
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia
| | - V I Stashkiv
- Vishnevsky National Medical Research Center of Surgery, Moscow, Russia
| |
Collapse
|
14
|
Dickinson SM, McIntyre CA, Schilsky JB, Harrington KA, Gerst SR, Flynn JR, Gonen M, Capanu M, Wong W, Lawrence S, Allen PJ, O'Reilly EM, Jarnagin WR, D'Angelica MI, Balachandran VP, Drebin JA, Kingham TP, Simpson AL, Do RK. Preoperative CT predictors of survival in patients with pancreatic ductal adenocarcinoma undergoing curative intent surgery. Abdom Radiol (NY) 2021; 46:1607-1617. [PMID: 32986175 PMCID: PMC8004545 DOI: 10.1007/s00261-020-02726-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/16/2020] [Accepted: 08/30/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To evaluate the associations between computed tomography (CT) imaging features extracted from the structured American Pancreatic Association (APA)/Society of Abdominal Radiology (SAR) template and overall survival in patients with resected pancreatic ductal adenocarcinoma (PDAC). METHODS This retrospective analysis included consecutive patients with PDAC who consented to genomic tumor testing and underwent preoperative imaging and curative intent surgical resection from December 2006 to July 2017. Two radiologists assessed preoperative CT imaging using the APA/SAR PDAC-reporting template. Univariable associations between overall survival and imaging variables were evaluated using Cox proportional hazards regression. RESULTS The study included 168 patients (66 years ± 11; 91 women). 126/168 patients (75%) received upfront surgical resection whereas 42/168 (25%) received neoadjuvant therapy prior to surgical resection. In the entire cohort, features associated with decreased overall survival were tumor arterial contact of any kind (hazard ratio (HR) 1.89, 95% CI 1.13-3.14, p = 0.020), tumor contact with the common hepatic artery (HR 2.33, 95% CI 1.35-4.04, p = 0.009), and portal vein deformity (HR 3.22, 95% CI 1.63-6.37, p = 0.003). In the upfront surgical group, larger tumor size was associated with decreased overall survival (HR 2.30, 95% CI 1.19-4.42, p = 0.013). In the neoadjuvant therapy group, the presence of venous collaterals was the only feature associated with decreased overall survival (HR 2.28, 95% CI 1.04-4.99, p = 0.042). CONCLUSION The application of the APA/SAR pancreatic adenocarcinoma reporting template may identify predictors of survival that can aid in preoperative stratification of patients.
Collapse
Affiliation(s)
- Shannan M Dickinson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Caitlin A McIntyre
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana B Schilsky
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Kate A Harrington
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Scott R Gerst
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Jessica R Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gonen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Winston Wong
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sharon Lawrence
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Peter J Allen
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William R Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael I D'Angelica
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vinod P Balachandran
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeffrey A Drebin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - T Peter Kingham
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amber L Simpson
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Biomedical and Molecular Sciences, School of Medicine, Queen's University, Kingston, ON, Canada
| | - Richard K Do
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| |
Collapse
|
15
|
Zaid M, Widmann L, Dai A, Sun K, Zhang J, Zhao J, Hurd MW, Varadhachary GR, Wolff RA, Maitra A, Katz MHG, Herman JM, Wang H, Knopp MV, Williams TM, Bhosale P, Tamm EP, Koay EJ. Predictive Modeling for Voxel-Based Quantification of Imaging-Based Subtypes of Pancreatic Ductal Adenocarcinoma (PDAC): A Multi-Institutional Study. Cancers (Basel) 2020; 12:E3656. [PMID: 33291471 PMCID: PMC7762105 DOI: 10.3390/cancers12123656] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 01/19/2023] Open
Abstract
Previously, we characterized qualitative imaging-based subtypes of pancreatic ductal adenocarcinoma (PDAC) on computed tomography (CT) scans. Conspicuous (high delta) PDAC tumors are more likely to have aggressive biology and poorer clinical outcomes compared to inconspicuous (low delta) tumors. Here, we developed a quantitative classification of this imaging-based subtype (quantitative delta; q-delta). Retrospectively, baseline pancreatic protocol CT scans of three cohorts (cohort#1 = 101, cohort#2 = 90 and cohort#3 = 16 [external validation]) of patients with PDAC were qualitatively classified into high and low delta. We used a voxel-based method to volumetrically quantify tumor enhancement while referencing normal-pancreatic-parenchyma and used machine learning-based analysis to build a predictive model. In addition, we quantified the stromal content using hematoxylin- and eosin-stained treatment-naïve PDAC sections. Analyses revealed that PDAC quantitative enhancement values are predictive of the qualitative delta scoring and were used to build a classification model (q-delta). Compared to high q-delta, low q-delta tumors were associated with improved outcomes, and the q-delta class was an independent prognostic factor for survival. In addition, low q-delta tumors had higher stromal content and lower cellularity compared to high q-delta tumors. Our results suggest that q-delta classification provides a clinically and biologically relevant tool that may be integrated into ongoing and future clinical trials.
Collapse
Affiliation(s)
- Mohamed Zaid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Lauren Widmann
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Annie Dai
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Kevin Sun
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Jun Zhao
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.Z.); (M.W.H.)
| | - Mark W. Hurd
- Sheikh Ahmed Center for Pancreatic Cancer Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (J.Z.); (M.W.H.)
| | - Gauri R. Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.R.V.); (R.A.W.)
| | - Robert A. Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (G.R.V.); (R.A.W.)
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
| | - Matthew H. G. Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Joseph M. Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.M.); (H.W.)
| | - Michael V. Knopp
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Terence M. Williams
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA;
| | - Priya Bhosale
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.P.T.)
| | - Eric P. Tamm
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (P.B.); (E.P.T.)
| | - Eugene J. Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (M.Z.); (L.W.); (A.D.); (K.S.); (J.M.H.)
| |
Collapse
|
16
|
Bartoli M, Barat M, Dohan A, Gaujoux S, Coriat R, Hoeffel C, Cassinotto C, Chassagnon G, Soyer P. CT and MRI of pancreatic tumors: an update in the era of radiomics. Jpn J Radiol 2020; 38:1111-1124. [PMID: 33085029 DOI: 10.1007/s11604-020-01057-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
Abstract
Radiomics is a relatively new approach for image analysis. As a part of radiomics, texture analysis, which consists in extracting a great amount of quantitative data from original images, can be used to identify specific features that can help determining the actual nature of a pancreatic lesion and providing other information such as resectability, tumor grade, tumor response to neoadjuvant therapy or survival after surgery. In this review, the basic of radiomics, recent developments and the results of texture analysis using computed tomography and magnetic resonance imaging in the field of pancreatic tumors are presented. Future applications of radiomics, such as artificial intelligence, are discussed.
Collapse
Affiliation(s)
- Marion Bartoli
- Department of Radiology, Cochin Hospital, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Maxime Barat
- Department of Radiology, Cochin Hospital, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France
| | - Anthony Dohan
- Department of Radiology, Cochin Hospital, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France
| | - Sébastien Gaujoux
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France
- Department of Abdominal Surgery, Cochin Hospital, AP-HP, 75014, Paris, France
| | - Romain Coriat
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France
- Department of Gastroenterology, Cochin Hospital, AP-HP, 75014, Paris, France
| | - Christine Hoeffel
- Department of Radiology, Robert Debré Hospital, 51092, Reims, France
| | - Christophe Cassinotto
- Department of Radiology, CHU Montpellier, University of Montpellier, Saint-Éloi Hospital, 34000, Montpellier, France
| | - Guillaume Chassagnon
- Department of Radiology, Cochin Hospital, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France
| | - Philippe Soyer
- Department of Radiology, Cochin Hospital, AP-HP, 27 Rue du Faubourg Saint-Jacques, 75014, Paris, France.
- Université de Paris, Descartes-Paris 5, F-75006, Paris, France.
| |
Collapse
|
17
|
Rhee H, Park MS. The Role of Imaging in Current Treatment Strategies for Pancreatic Adenocarcinoma. Korean J Radiol 2020; 22:23-40. [PMID: 32901458 PMCID: PMC7772381 DOI: 10.3348/kjr.2019.0862] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 04/30/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023] Open
Abstract
In pancreatic cancer, imaging plays an essential role in surveillance, diagnosis, resectability evaluation, and treatment response evaluation. Pancreatic cancer surveillance in high-risk individuals has been attempted using endoscopic ultrasound (EUS) or magnetic resonance imaging (MRI). Imaging diagnosis and resectability evaluation are the most important factors influencing treatment decisions, where computed tomography (CT) is the preferred modality. EUS, MRI, and positron emission tomography play a complementary role to CT. Treatment response evaluation is of increasing clinical importance, especially in patients undergoing neoadjuvant therapy. This review aimed to comprehensively review the role of imaging in relation to the current treatment strategy for pancreatic cancer, including surveillance, diagnosis, evaluation of resectability and treatment response, and prediction of prognosis.
Collapse
Affiliation(s)
- Hyungjin Rhee
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Mi Suk Park
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Seo W, Kim YC, Min SJ, Lee SM. Enhancement parameters of contrast-enhanced computed tomography for pancreatic ductal adenocarcinoma: Correlation with pathologic grading. World J Gastroenterol 2020; 26:4151-4158. [PMID: 32821076 PMCID: PMC7403799 DOI: 10.3748/wjg.v26.i28.4151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 05/08/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDA) is a malignancy with a high mortality rate and short survival time. The conventional computed tomography (CT) has been worldwide used as a modality for diagnosis of PDA, as CT enhancement pattern has been thought to be related to tumor angiogenesis and pathologic grade of PDA.
AIM To evaluate the relationship between the pathologic grade of pancreatic ductal adenocarcinoma and the enhancement parameters of contrast-enhanced CT.
METHODS In this retrospective study, 42 patients (Age, mean ± SD: 62.43 ± 11.42 years) with PDA who underwent surgery after preoperative CT were selected. Two radiologists evaluated the CT images and calculated the value of attenuation at the aorta in the arterial phase and the pancreatic phase (VAarterial and VApancreatic) and of the tumor (VTarterial and VTpancreatic) by finding out four regions of interest. Ratio between the tumor and the aorta enhancement on the arterial phase and the pancreatic phase (TARarterial and TARpancreatic) was figured out through dividing VTarterial by VAarterial and VTpancreatic by VApancreatic. Tumor-to-aortic enhancement fraction (TAF) was expressed as the ratio of the difference between attenuation of the tumor on arterial and parenchymal images to that between attenuation of the aorta on arterial and pancreatic images. The Kruskal-Wallis analysis of variance and Mann-Whitney U test for statistical analysis were used.
RESULTS Forty-two PDAs (23 men and 19 women) were divided into three groups: Well-differentiated (n = 13), moderately differentiated (n = 21), and poorly differentiated (n = 8). TAF differed significantly between the three groups (P = 0.034) but TARarterial (P = 0.164) and TARpancreatic (P = 0.339) did not. The median value of TAF for poorly differentiated PDAs (0.1011; 95%CI: 0.01100-0.1796) was significantly higher than that for well-differentiated PDAs (0.1941; 95%CI: 0.1463-0.3194).
CONCLUSION Calculation of TAF might be useful in predicting the pathologic grade of PDA.
Collapse
Affiliation(s)
- Woorim Seo
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Gyeonggi-do 18450, South Korea
| | - Young Chul Kim
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Gyeonggi-do 18450, South Korea
| | - Seon Jeong Min
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Gyeonggi-do 18450, South Korea
| | - Sang Min Lee
- Department of Radiology, Hallym University Sacred Heart Hospital, Gyeonggi-do 14068, South Korea
| |
Collapse
|
19
|
Pancreatic adenocarcinoma: quantitative CT features are correlated with fibrous stromal fraction and help predict outcome after resection. Eur Radiol 2020; 30:5158-5169. [PMID: 32346792 DOI: 10.1007/s00330-020-06853-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 03/09/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To identify quantitative imaging features of contrast-enhanced computed tomography (CE-CT) that may be prognostically favorable after resection of smaller (≤ 30 mm) pancreatic ductal adenocarcinomas (PDACs) located at head. METHODS This retrospective study included two independent cohorts (discovery cohort, n = 212; test cohort, n = 100) of patients who underwent resection of head PDACs ≤ 30 mm and preoperative CE-CT. We examined tumor and surrounding parenchymal attenuation differences (deltas), and tumor attenuation changes across phases (ratios). Semantic features of PDACs were evaluated by two radiologists. Clinicopathologic and imaging features for predicting disease-free survival (DFS) and overall survival (OS) were analyzed via multivariate Lasso-penalized Cox proportional-hazards models. Survival rates were derived by Kaplan-Meier method. RESULTS Imaging features achieved C-indices of 0.766 (discovery cohort) and 0.739 (test cohort) for DFS, and 0.790 (discovery cohort) and 0.772 (test cohort) for OS estimates through incorporation of clinicopathologic features. The most decisive imaging feature was delta 3, denoting attenuation differences between tumor and surrounding pancreas at pancreatic phase (DFS: HR = 2.122; OS: HR = 2.375; both p < 0.001). Compared with inconspicuous (low-delta-3, < 28 HU) tumors, conspicuous (high-delta-3) tumors correlated significantly with more aggressive histologic grades (p = 0.014) and less extensive tumor fibrous stromal fractions (p < 0.001). Patients with low-delta-3 tumors ≤ 20 mm experienced the most favorable outcomes (DFS, 36 months; OS, 42 months), whereas those with high-delta-3 tumors fared poorly, regardless of tumor size (DFS, 12 months; OS, 19 months). CONCLUSIONS Quantifiable CT imaging features reflect heterogeneous fibrous stromal fractions and histologic grades of PDAC at head locations that help stratify patients with disparate clinical outcomes. KEY POINTS • Quantitative and semantic imaging features achieved promising results for the prognosis of resected PDAC (≤ 30 mm) at head location, through incorporation of clinicopathologic features. • Attenuation difference at tumor-parenchyma interface (delta 3) emerged as the most decisive imaging feature, enabling further stratification of patients into distinct prognostic subtypes by tumor size. • High delta 3 signifies sharper contrast between tumor and surrounding pancreas, correlating with more aggressive histologic grades and less extensive tumor fibrous stromal fractions.
Collapse
|
20
|
Meng X, Liu P, Wu Y, Liu X, Huang Y, Yu B, Han J, Jin H, Tan X. Integrin beta 4 (ITGB4) and its tyrosine-1510 phosphorylation promote pancreatic tumorigenesis and regulate the MEK1-ERK1/2 signaling pathway. Bosn J Basic Med Sci 2020; 20:106-116. [PMID: 31242404 PMCID: PMC7029197 DOI: 10.17305/bjbms.2019.4255] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death, with a 5-year survival rate of only 1–4%. Integrin-mediated cell adhesion is critical for the initiation, progression, and metastasis of cancer. In this study we investigated the role of integrin β4 (ITGB4) and its phosphorylation at tyrosine Y1510 (p-ITGB4-Y1510) in the tumorigenesis of pancreatic cancer. We analyzed the expression of ITGB4 and p-ITGB4-Y1510 in pancreatic cancer tissue and cell lines using immunohistochemistry, Western blot, or semi-quantitative reverse transcription PCR. ITGB4 and p-ITGB4-Y1510 were highly expressed in pancreatic cancer (n = 176) compared with normal pancreatic tissue (n = 171). High p-ITGB4-Y1510 expression correlated with local invasion and distant metastasis of pancreatic cancer, and high ITGB4 was significantly associated with poor survival of patients. Inhibition of ITGB4 by siRNA significantly reduced migration and invasion of PC-1.0 and AsPC-1 cells. Overexpression of the mutant ITGB4-Y1510A (a mutation of tyrosine to alanine at 1510 position) in PC-1.0 and AsPC-1 cells not only blocked the ITGB4 phosphorylation at Y1510 but also suppressed the expression of ITGB4 (p < 0.05 vs. wild-type ITGB4). The transfection of PC-1.0 and AsPC-1 cells with ITGB4-Y1510A significantly decreased the level of p-mitogen-activated protein kinase kinase (MEK)1 (T292) and p-extracellular signal-regulated kinase (ERK)1/2 but did not affect the level of p-MEK1 (T386) and p-MEK2 (T394). Overall, our study showed that ITGB4 and its phosphorylated form promote cell migration and invasion in pancreatic cancer and that p-ITGB4-Y1510 regulates the downstream MEK1-ERK1/2 signaling cascades. Targeting ITGB4 or its phosphorylation at Y1510 may be a novel therapeutic option for pancreatic cancer.
Collapse
Affiliation(s)
- Xiangli Meng
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Peng Liu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Yunhao Wu
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China
| | - Xinlu Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yinpeng Huang
- Minimally Invasive Area of General Surgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Boqiang Yu
- Department of General Surgery, Fushun Central Hospital, Fushun, China
| | - Jiahong Han
- Department of Surgery, Liaoning Electric Power Center Hospital, Shenyang, China.
| | - Haoyi Jin
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| | - Xiaodong Tan
- Department of the First General Surgery, Shengjing Hospital affiliated to China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Sandini M, Negreros-Osuna AA, Qadan M, Hank T, Patino M, Ferrone CR, Warshaw AL, Lillemoe KD, Sahani D, Castillo CFD. Main Pancreatic Duct to Parenchymal Thickness Ratio at Preoperative Imaging is Associated with Overall Survival in Upfront Resected Pancreatic Cancer. Ann Surg Oncol 2019; 27:1606-1612. [PMID: 31722071 DOI: 10.1245/s10434-019-08040-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic cancer induces parenchymal atrophy and duct dilation. The aim of this study was to evaluate whether these radiologic modifications are associated with outcomes. METHODS Upfront pancreaticoduodenectomy patients with available preoperative contrast enhanced CT scan imaging were retrospectively analyzed. Thickness of the pancreas, size of the main pancreatic duct (MPD), and distance of the tumor from the ampulla were assessed. A training cohort was selected, including short- (3-12 months following surgery) and long-term (≥ 36 months) survivors. Identified survival determinants were validated in the overall cohort. RESULTS Two-hundred-sixteen patients were analyzed. In the training cohort (N = 118), 68 patients (57.6%) were in the short-term and 50 (42.4%) in the long-term survival group. The short-term survival group had significantly higher CA 19-9 levels (p = 0.027), larger tumors (32.6 ± 12.1 mm vs. 26.5 ± 11.6 mm, p = 0.007), poorer differentiation (p = 0.003), higher rate of R < 1 mm resections (54% vs. 32%, p = 0.008), and reduced receipt of adjuvant chemotherapy (p = 0.020). The MPD-to-pancreatic thickness ratio was significantly lower in the short-term survivors (3.6 ± 6.2 vs. 8.2 ± 12.0, p = 0.016). In the entire cohort, an MPD-to-pancreatic thickness ratio ≥ 3.5 was associated with improved OS [median 33.0 months IQR (19.7-48.1) versus 17 months IQR (14.8-19.2), p = 0.004], and confirmed by a Cox-proportional hazards model independently associated with OS (HR = 0.58; p = 0.009), together with tumor size (HR = 1.02; p =0.012), R1/R2 status (HR = 1.53; p = 0.029), and receipt of adjuvant treatment (HR = 0.61; p = 0.021). CONCLUSIONS High MPD-to-pancreatic thickness ratio was associated with improved long-term survival in pancreaticoduodenectomy for cancer. Whether these features are related to tumor chronicity, indolent biology, or local growth over metastasis remains to be determined.
Collapse
Affiliation(s)
- Marta Sandini
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Adrian A Negreros-Osuna
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Hank
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Manuel Patino
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew L Warshaw
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dushyant Sahani
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
22
|
MDCT findings predicting post-operative residual tumor and survival in patients with pancreatic cancer. Eur Radiol 2019; 29:3714-3724. [PMID: 30899975 DOI: 10.1007/s00330-019-06140-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/15/2019] [Accepted: 03/08/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To predict residual tumor (R) classification and overall survival (OS) on preoperative MDCT in patients who underwent first-line surgery for pancreatic ductal adenocarcinoma (PDA). METHODS Three hundred sixteen patients with PDA who underwent MDCT and first-line surgery were included. Patients were divided into a test (n = 216) and a validation group (n = 100). The R classification was categorized into R0 (no residual tumor) and R1/R2 (microscopic/macroscopic residual tumor). We assessed the correlation between the MDCT findings and the R classification. For survival analysis, we used the Kaplan-Meier estimation and Cox proportional hazard model to determine the prognostic factors for OS. Validation of the prediction models for the R classification and OS was performed using C statistics and calibration plot. RESULTS Peritumoral fat stranding (odds ratio (OR) 3.826), suspicious distant metastasis (OR 2.916), portal vein involvement (OR 2.795), and tumor size (OR 1.045) were independent predictors for residual tumor (p < .05). On survival analysis, common hepatic artery involvement (hazard ratio (HR) 5.656), R1/R2 stage (HR 2.476), and N1 stage (HR 1.745) were predictors of poor OS (p < .05). C statistics for prediction models for R classification and OS were 0.816 and 0.662, respectively. Calibration plots showed good predictive performance in a high probability of the R1/R2 stage or poor OS. CONCLUSION Preoperative MDCT is useful for predicting the R classification using the tumor size, peritumoral fat stranding, portal vein involvement, and suspicious distant metastasis, as well as for anticipating poor OS using the N1 stage, common hepatic artery involvement, and R1/R2 stage in patients with PDA. KEY POINTS • Thorough assessment of the involvement of common hepatic artery or portal vein and peritumoral fat stranding is warranted for predicting prognosis in patients with pancreatic ductal adenocarcinoma. • Not only encasement but also abutment of common hepatic artery or portal vein by tumor predicts poor prognosis after upfront surgery. • If residual tumor or poor overall survival is anticipated on preoperative MDCT, neoadjuvant treatment can be performed.
Collapse
|
23
|
Lee S, Kim SH, Park HK, Jang KT, Hwang JA, Kim S. Pancreatic Ductal Adenocarcinoma: Rim Enhancement at MR Imaging Predicts Prognosis after Curative Resection. Radiology 2018; 288:456-466. [DOI: 10.1148/radiol.2018172331] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Sunyoung Lee
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| | - Seong Hyun Kim
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| | - Hyung Kyu Park
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| | - Kee Taek Jang
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| | - Jeong Ah Hwang
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| | - Seonwoo Kim
- From the Department of Radiology and Center for Imaging Science (S.L., S.H.K., J.A.H.), Department of Pathology (H.K.P., K.T.J.), and Department of Statistics and Data Center (S.K.), Samsung Medical Center, Sungkyunkwan University School of Medicine, 81 Irwon-Ro, Gangnam-gu, Seoul 06351, Korea
| |
Collapse
|
24
|
Kang MJ, Jang JY, Kwon W, Kim SW. Clinical significance of defining borderline resectable pancreatic cancer. Pancreatology 2018; 18:139-145. [PMID: 29274720 DOI: 10.1016/j.pan.2017.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/11/2022]
Abstract
Since the introduction of the concept of borderline resectable pancreatic cancer (BRPC), various definitions of this disease entity have been suggested. However, there are several obstacles in defining this disease category. The current diagnostic criteria of BRPC mainly focuses on its expanded 'technical resectability'; however, they are difficult to interpret because of their ambiguity using potential subjective or arbitrary terminology, In addition, limitations in current imaging technology and a lack of evidence in radiological-pathological-clinical correlation make it difficult to refine the criteria. On the other hand, neoadjuvant treatment is usually applied to increase the R0 resection rate of BRPC focusing on the 'oncological curability'. However, evidence is needed concerning the effect of neoadjuvant treatment by quality-controlled prospective randomized clinical trials based on a standardized radiologic and pathologic reporting system. In conclusion, there are two aspects in the current concept of BRPC, which are technical resectability and oncological curability. Although the recent evolution of surgical techniques is expanding the scope of technical resectability, it should not be overlooked that the disease entity must be defined based on the evidence of oncological curability.
Collapse
Affiliation(s)
- Mee Joo Kang
- Korea International Cooperation Agency, Republic of Korea
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sun-Whe Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Amer AM, Zaid M, Chaudhury B, Elganainy D, Lee Y, Wilke CT, Cloyd J, Wang H, Maitra A, Wolff RA, Varadhachary G, Overman MJ, Lee JE, Fleming JB, Tzeng CW, Katz MH, Holliday EB, Krishnan S, Minsky BD, Herman JM, Taniguchi CM, Das P, Crane CH, Le O, Bhosale P, Tamm EP, Koay EJ. Imaging-based biomarkers: Changes in the tumor interface of pancreatic ductal adenocarcinoma on computed tomography scans indicate response to cytotoxic therapy. Cancer 2018; 124:1701-1709. [PMID: 29370450 PMCID: PMC5891375 DOI: 10.1002/cncr.31251] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/22/2017] [Accepted: 12/21/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The assessment of pancreatic ductal adenocarcinoma (PDAC) response to therapy remains challenging. The objective of this study was to investigate whether changes in the tumor/parenchyma interface are associated with response. METHODS Computed tomography (CT) scans before and after therapy were reviewed in 4 cohorts: cohort 1 (99 patients with stage I/II PDAC who received neoadjuvant chemoradiation and surgery); cohort 2 (86 patients with stage IV PDAC who received chemotherapy), cohort 3 (94 patients with stage I/II PDAC who received protocol‐based neoadjuvant gemcitabine chemoradiation), and cohort 4 (47 patients with stage I/II PDAC who received neoadjuvant chemoradiation and were prospectively followed in a registry). The tumor/parenchyma interface was visually classified as either a type I response (the interface remained or became well defined) or a type II response (the interface became poorly defined) after therapy. Consensus (cohorts 1‐3) and individual (cohort 4) visual scoring was performed. Changes in enhancement at the interface were quantified using a proprietary platform. RESULTS In cohort 1, type I responders had a greater probability of achieving a complete or near‐complete pathologic response (21% vs 0%; P = .01). For cohorts 1, 2, and 3, type I responders had significantly longer disease‐free and overall survival, independent of traditional covariates of outcomes and of baseline and normalized cancer antigen 19‐9 levels. In cohort 4, 2 senior radiologists achieved a κ value of 0.8, and the interface score was associated with overall survival. The quantitative method revealed high specificity and sensitivity in classifying patients as type I or type II responders (with an area under the receiver operating curve of 0.92 in cohort 1, 0.96 in cohort 2, and 0.89 in cohort 3). CONCLUSIONS Changes at the PDAC/parenchyma interface may serve as an early predictor of response to therapy. Cancer 2018;124:1701‐9. © 2018 The Authors. Cancer published by Wiley Periodicals, Inc. on behalf of American Cancer Society. This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial‐NoDerivs License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made. An imaging feature of pancreatic cancer is identified that indicates a response to cytotoxic therapies. This may be helpful as an early predictor of response for clinical trials and for deciding whether to change therapy.
Collapse
Affiliation(s)
- Ahmed M Amer
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mohamed Zaid
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Baishali Chaudhury
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Dalia Elganainy
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yeonju Lee
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher T Wilke
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jordan Cloyd
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gauri Varadhachary
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael J Overman
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffery E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa Bay, Florida
| | - Ching Wei Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emma B Holliday
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bruce D Minsky
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph M Herman
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christopher H Crane
- Department of Radiation Oncology, Memorial Sloan Cancer Center, New York, New York
| | - Ott Le
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Priya Bhosale
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eric P Tamm
- Department of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
26
|
Novel Diagnostic and Predictive Biomarkers in Pancreatic Adenocarcinoma. Int J Mol Sci 2017; 18:ijms18030667. [PMID: 28335509 PMCID: PMC5372679 DOI: 10.3390/ijms18030667] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal disease for a multitude of reasons including very late diagnosis. This in part is due to the lack of understanding of the biological behavior of PDAC and the ineffective screening for this disease. Significant efforts have been dedicated to finding the appropriate serum and imaging biomarkers to help early detection and predict response to treatment of PDAC. Carbohydrate antigen 19-9 (CA 19-9) has been the most validated serum marker and has the highest positive predictive value as a stand-alone marker. When combined with carcinoembryonic antigen (CEA) and carbohydrate antigen 125 (CA 125), CA 19-9 can help predict the outcome of patients to surgery and chemotherapy. A slew of novel serum markers including multimarker panels as well as genetic and epigenetic materials have potential for early detection of pancreatic cancer, although these remain to be validated in larger trials. Imaging studies may not correlate with elevated serum markers. Critical features for determining PDAC include the presence of a mass, dilated pancreatic duct, and a duct cut-off sign. Features that are indicative of early metastasis includes neurovascular bundle involvement, duodenal invasion, and greater post contrast enhancement. 18-F-fluorodeoxyglucose (18-FDG) radiotracer uptake and changes following treatment may predict patient overall survival following treatment. Similarly, pretreatment apparent diffusion coefficient (ADC) values may predict prognosis with lower ADC lesions having worse outcome. Although these markers have provided significant improvement in the care of pancreatic cancer patients, further advancements can be made with perhaps better combination of markers or discovery of unique marker(s) to pancreatic cancer.
Collapse
|
27
|
Cassinotto C, Chong J, Zogopoulos G, Reinhold C, Chiche L, Lafourcade JP, Cuggia A, Terrebonne E, Dohan A, Gallix B. Resectable pancreatic adenocarcinoma: Role of CT quantitative imaging biomarkers for predicting pathology and patient outcomes. Eur J Radiol 2017; 90:152-158. [PMID: 28583627 DOI: 10.1016/j.ejrad.2017.02.033] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 02/18/2017] [Accepted: 02/20/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUNDS Patients with a pancreatic cancer amenable to surgery still have a poor prognosis and high risk of post-operative recurrence. We aimed to assess the value of quantitative imaging biomarkers using computed-tomography (CT) texture analysis to evaluate the pathologic tumor aggressiveness and predict disease-free survival (DFS) in patients with resectable pancreatic adenocarcinoma. METHODS We retrospectively performed attenuation measurements and texture analysis on the portal-venous phase of the pre-operative CT scan of 99 patients that underwent resection of a pancreatic ductal adenocarcinoma in two university hospitals. Tumor attenuation parameters included: mean attenuation value of the whole tumor (WHOLE-AV), and of the most hypoattenuating area within the tumor (CENTRAL-AV). Tumor heterogeneity parameters included: standard deviation, entropy, skewness, and kurtosis. RESULTS Tumor attenuation parameters showed significant association with the tumor differentiation grade (CENTRAL-AV, Odds ratio (OR) 0.968, 95% confidence interval (CI) 0.94-0.998) and lymph node invasion (WHOLE-AV, OR 0.886, CI 0.823-0.955). Variables associated with early-recurrence were: lymph node ratio (R2=0.15), kurtosis (R2=0.08), and CENTRAL-AV (R2=0.04). Lymph node ratio (Hazard ratio (HR) 1.02), and CENTRAL-AV (HR 0.98) were independently associated with shorter DFS. Patients with CENTRAL-AV<62 Hounsfield units had a shorter 1-year DFS (35% versus 68%, p=0.004). CONCLUSION Tumors that are more hypoattenuating on the portal-venous phase on CT scan are potentially more aggressive with higher tumor grade, greater lymph node invasion, and shorter DFS.
Collapse
Affiliation(s)
- Christophe Cassinotto
- Department of Radiology, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - Jaron Chong
- Department of Radiology, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - George Zogopoulos
- Department of Visceral Surgery, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - Caroline Reinhold
- Department of Radiology, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - Laurence Chiche
- Department of Visceral Surgery, Hôpital Haut-Lévêque, University Hospital of Bordeaux, 1 Avenue de Magellan, 33604 Pessac, France.
| | - Jean-Pierre Lafourcade
- Department of Diagnostic and Interventional Radiology, Hôpital Haut-Lévêque, University Hospital of Bordeaux, 1 avenue de Magellan, 33604 Pessac, France.
| | - Adeline Cuggia
- Department of Visceral Surgery, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - Eric Terrebonne
- Department of Digestive Oncology, Hôpital Haut-Lévêque, University Hospital of Bordeaux, 1 Avenue de Magellan, 33604 Pessac, France.
| | - Anthony Dohan
- Department of Radiology, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| | - Benoît Gallix
- Department of Radiology, Royal Victoria Hospital, McGill University Health Center, 1001 Boulevard Decarie, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
28
|
Erratum: CT Imaging Biomarkers Predict Clinical Outcomes After Pancreatic Cancer Surgery: Erratum. Medicine (Baltimore) 2016; 95:e29ce. [PMID: 31265559 PMCID: PMC4839788 DOI: 10.1097/01.md.0000482791.58629.ce] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
[This corrects the article DOI: 10.1097/MD.0000000000002664.].
Collapse
|